1
|
Mao S, Li J, Huang J, Lv L, Zhang Q, Cheng Q, Liu X, Bi Z, Yao J. Therapeutic potential of microRNA-506 in cancer treatment: mechanisms and therapeutic implications. Front Oncol 2025; 15:1524763. [PMID: 40248198 PMCID: PMC12003368 DOI: 10.3389/fonc.2025.1524763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 03/17/2025] [Indexed: 04/19/2025] Open
Abstract
Cancer is a complex and highly lethal disease marked by unchecked cell proliferation, aggressive behavior, and a strong tendency to metastasize. Despite significant advancements in cancer diagnosis and treatment, challenges such as early detection difficulties, drug resistance, and adverse effects of radiotherapy or chemotherapy continue to threaten patient survival. MicroRNAs (miRNAs) have emerged as critical regulators in cancer biology, with miR-506 being extensively studied and recognized for its tumor-suppressive effects across multiple cancer types. This review examines the regulatory mechanisms of miR-506 in common cancers, focusing on its role in the competing endogenous RNA (ceRNA) network and its effects on cancer cell proliferation, apoptosis, and migration. We also discuss the potential of miR-506 as a therapeutic target and its role in overcoming drug resistance in cancer treatment. Overall, these insights underscore the therapeutic potential of miR-506 and its promise in developing novel cancer therapies.
Collapse
Affiliation(s)
- Shuzhen Mao
- Department of Pharmacy, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Junyan Li
- Department of Pathology, Second People’s Hospital of Ningyang, Taian, Shandong, China
| | - Jiahui Huang
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Lili Lv
- Department of Pathology, Second People’s Hospital of Ningyang, Taian, Shandong, China
| | - Qilian Zhang
- Department of Pathology, People’s Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Qing Cheng
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Xiaojing Liu
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Zhiwei Bi
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Jing Yao
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| |
Collapse
|
2
|
Wei Y, Wang P, Zhao J, Fan X, Jiang J, Mu X, Wang Y, Yang A, Zhang R, Hu S, Guo Z. Overexpression of miR-124 enhances the therapeutic benefit of TMZ treatment in the orthotopic GBM mice model by inhibition of DNA damage repair. Cell Death Dis 2025; 16:47. [PMID: 39865088 PMCID: PMC11770086 DOI: 10.1038/s41419-025-07363-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 12/20/2024] [Accepted: 01/16/2025] [Indexed: 01/28/2025]
Abstract
Glioblastoma (GBM) is the most common malignant primary brain cancer with poor prognosis due to the resistant to current treatments, including the first-line drug temozolomide (TMZ). Accordingly, it is urgent to clarify the mechanism of chemotherapeutic resistance to improve the survival rate of patients. In the present study, by integrating comprehensive non-coding RNA-seq data from multiple cohorts of GBM patients, we identified that a series of miRNAs are frequently downregulated in GBM patients compared with the control samples. Among them, a high level of miR-124 is closely associated with a favorable survival rate in the clinical patients. In the phenotype experiment, we demonstrated that miR-124 overexpression increases responsiveness of GBM cells to TMZ-induced cell death, and vice versa. In the mechanistic study, we for the first time identified that RAD51, a key functional molecule in DNA damage repair, is a novel and bona fide target of miR-124 in GBM cells. Given that other miR-124-regulated mechanisms on TMZ sensitivity have been reported, we performed recue experiment to demonstrate that RAD51 is essential for miR-124-mediated sensitivity to TMZ in GBM cells. More importantly, our in vivo functional experiment showed that combinational utilization of miR-124 overexpression and TMZ presents a synergetic therapeutic benefit in the orthotopic GBM mice model. Taken together, we rationally explained a novel and important mechanism of the miR-124-mediated high sensitivity to TMZ-induced cell death in GBM and provided evidence to support that miR-124-RAD51 regulatory axis could be a promising candidate in the comprehensive treatment with TMZ in GBM.
Collapse
Affiliation(s)
- Yuchen Wei
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Peng Wang
- Department of Neurosurgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jianhui Zhao
- Department of Critical Care Medicine, Hainan Hospital of Chinese PLA General Hospital, Sanya City, Hainan Province, China
| | - Xin Fan
- Department of Otolaryngology Head and Neck Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Jun Jiang
- Department of Health Service, Base of Health Service, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Xiuli Mu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Yuzhou Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Angang Yang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Rui Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi Province, China.
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi Province, China.
| | - Shijie Hu
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China.
| | - Zhangyan Guo
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi Province, China.
| |
Collapse
|
3
|
Jia C, Liu Q, Zhang M, Han C, Luo X, Zhou Y, Liu Y, Zhang L. Engineering Nano-Pills to Inhibit Ovarian Cancer Proliferation and Migration through a Combination of Chemical/Nucleic Acid Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2408095. [PMID: 39575476 DOI: 10.1002/smll.202408095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/19/2024] [Indexed: 01/23/2025]
Abstract
Ovarian cancer (OC) is the most fatal of all gynecological malignancies, presenting a significant threat to women's health. Its treatment is complicated by severe dose-dependent chemotherapy toxicity, drug resistance, and tumor migration. Herein, an intelligent combination strategy of chemotherapy and nucleic acid therapy, named ApMEmiR&D is developed. This integrated system consists of three parts: the nano-pill, the protective membrane, and the navigation element. Nano-pills are nanospheres assembled from miRNA and doxorubicin (DOX) with the help of ferrous ions (Fe2+). The protective membrane is derived from tumor-associated macrophages (TAMs membrane) originating from the primary tumor microenvironment (TME). The navigation element is the cholesterol-conjugated AS1411 aptamer. The resulting ApMEmiR&D nanoparticles exhibit uniform size, a well-defined nanosphere structure, robust serum stability, and ultra-high drug loading efficiency and capacity. The system can efficiently accumulate in the tumor, allowing for the synergistic inhibition of tumor growth and metastasis without apparent systemic toxicity. The results demonstrate the homing effect of tumor microenvironment-derived macrophage cell membrane and the targeting effect of aptamer, leading to precise drug targeting and immune compatibility, thereby enhancing therapeutic efficacy. The success of this strategy paves the way for metastasis inhibition and targeted cancer therapy.
Collapse
Affiliation(s)
- Chao Jia
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, College of Pharmacy, Nankai University, Tianjin, 300350, P. R. China
- Nankai International Advanced Research Institute, Shenzhen, Guangdong, 518045, P. R. China
| | - Qirui Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, College of Pharmacy, Nankai University, Tianjin, 300350, P. R. China
| | - Man Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, College of Pharmacy, Nankai University, Tianjin, 300350, P. R. China
| | - Cong Han
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, College of Pharmacy, Nankai University, Tianjin, 300350, P. R. China
| | - Xuantong Luo
- Tianjin Haihe High School, Tianjin, 300202, P. R. China
| | - Yu Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, College of Pharmacy, Nankai University, Tianjin, 300350, P. R. China
| | - Yi Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, College of Pharmacy, Nankai University, Tianjin, 300350, P. R. China
| | - Liyun Zhang
- Nankai International Advanced Research Institute, Shenzhen, Guangdong, 518045, P. R. China
| |
Collapse
|
4
|
Pellegrino B, Capoluongo ED, Bagnoli M, Arenare L, Califano D, Scambia G, Cecere SC, Silini EM, Scaglione GL, Spina A, Tognon G, Campanini N, Pisano C, Russo D, Pettinato A, Scollo P, Iemmolo R, De Cecco L, Musolino A, Marchini S, Beltrame L, Paracchini L, Perrone F, Mezzanzanica D, Pignata S. Unraveling the complexity of HRD assessment in ovarian cancer by combining genomic and functional approaches: translational analyses of MITO16-MaNGO-OV-2 trial. ESMO Open 2025; 10:104091. [PMID: 39754985 PMCID: PMC11758122 DOI: 10.1016/j.esmoop.2024.104091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 11/26/2024] [Accepted: 11/26/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Ovarian cancer (OvC) constitutes significant management challenges primarily due to its late-stage diagnosis and the development of resistance to chemotherapy. The standard treatment regimen typically includes carboplatin and paclitaxel, with the addition of poly (ADP-ribose) polymerase inhibitors for patients with high-grade serous ovarian cancer (HGSOC) harboring BRCA1/2 mutations. However, the variability in treatment responses suggests the need to investigate factors beyond BRCA1/2 mutations, such as DNA repair mechanisms and epigenetic alterations. Notably, homologous recombination repair deficiency (HRD) is observed in an additional 20% of HGSOC cases, indicating a broader spectrum of DNA repair defects. Existing commercial HRD assays have certain limitations, prompting a global effort to develop new genomic and functional tests through academic research. MATERIALS AND METHODS This study investigates, in the 187 high-grade serous and endometrioid tumors from the MITO16/MaNGO-OV-2 trial, academic HRD genomic tests in conjunction with a RAD51 immunofluorescence assay to assess functional activation of HRD. Additionally, the study incorporates analysis of microRNA-506 (miR-506) expression as a putative epigenetic effector. RESULTS The RAD51 test identified HRD in 73% of the samples and genomic HRD testing in 57%, with HRD identified in 45% of samples by both tests. The significant discrepancy between the two assays emphasizes the need to refine tumor classification for HRD. A three-group genomic classification unveiled superior progression-free survival (PFS) in high- and mild-HRD tumors compared with negative-HRD tumors. High concordance between RAD51 and genomic testing in high-HRD tumors suggests a subset of 'super-HRD' tumors exhibiting superior PFS. High expression of miR-506 may be used to further refine HRD status. CONCLUSIONS The study underscores the complexities of HRD assessment and advocates for a combined genomic and functional approach to enhance predictive accuracy in OvC treatment responses.
Collapse
Affiliation(s)
- B Pellegrino
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy; Breast Unit, University Hospital of Parma, Parma, Italy
| | - E D Capoluongo
- Department of Molecular Medicine and Medical Biotechnology, Università degli Studi di Napoli Federico II, Naples, Italy; Department of Clinical Pathology, Azienda Ospedaliera San Giovanni Addolorata, Rome, Italy
| | - M Bagnoli
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - L Arenare
- Clinical Trial Unit, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Naples, Italy
| | - D Califano
- Microenvironment Molecular Targets Unit, Istituto Nazionale Tumori IRCCS e Fondazione G. Pascale, Naples, Italy
| | - G Scambia
- Gynecologic Oncology Unit, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy; Catholic University of Sacred Heart, Rome, Italy
| | - S C Cecere
- Uro-Gynecologic Oncology Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy
| | - E M Silini
- Unit of Pathological Anatomy, Department of Medicine and Surgery, University Hospital of Parma, Parma, Italy
| | - G L Scaglione
- Laboratory of Molecular Oncology, IDI-IRCCS, Rome, Italy
| | - A Spina
- Microenvironment Molecular Targets Unit, Istituto Nazionale Tumori IRCCS e Fondazione G. Pascale, Naples, Italy
| | - G Tognon
- Division of Obstetrics and Gynecology, ASST Spedali Civili di Brescia, Brescia, Italy
| | - N Campanini
- Unit of Pathological Anatomy, Department of Medicine and Surgery, University Hospital of Parma, Parma, Italy
| | - C Pisano
- Uro-Gynecologic Oncology Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy
| | - D Russo
- Microenvironment Molecular Targets Unit, Istituto Nazionale Tumori IRCCS e Fondazione G. Pascale, Naples, Italy
| | - A Pettinato
- Department of Pathological Anatomy, A.O.E. Cannizzaro, Catania, Italy
| | - P Scollo
- Division of Gynecology and Obstetrics, Maternal and Child Department, Cannizzaro Hospital, Kore University of Enna, Enna, Italy
| | - R Iemmolo
- Laboratory of Genomics, L.C. Laboratori Campisi, Avola, Italy
| | - L De Cecco
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - A Musolino
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy; Breast Unit, University Hospital of Parma, Parma, Italy; Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - S Marchini
- Cancer Pharmacology Lab, IRCCS Humanitas Research Hospital, Milan, Italy
| | - L Beltrame
- Cancer Pharmacology Lab, IRCCS Humanitas Research Hospital, Milan, Italy
| | - L Paracchini
- Cancer Pharmacology Lab, IRCCS Humanitas Research Hospital, Milan, Italy
| | - F Perrone
- Clinical Trial Unit, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Naples, Italy
| | - D Mezzanzanica
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - S Pignata
- Uro-Gynecologic Oncology Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy.
| |
Collapse
|
5
|
Dey Bhowmik A, Shaw P, Gopinatha Pillai MS, Rao G, Dwivedi SKD. Evolving landscape of detection and targeting miRNA/epigenetics for therapeutic strategies in ovarian cancer. Cancer Lett 2024; 611:217357. [PMID: 39615646 DOI: 10.1016/j.canlet.2024.217357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 12/14/2024]
Abstract
Ovarian cancer (OC) accounts for the highest mortality rates among all gynecologic malignancies. The high mortality of OC is often associated with delayed detection, prolonged latency, enhanced metastatic potential, acquired drug resistance, and frequent recurrence. This review comprehensively explores key aspects of OC, including cancer diagnosis, mechanisms of disease resistance, and the pivotal role of epigenetic regulation, particularly by microRNAs (miRs) in cancer progression. We highlight the intricate regulatory mechanisms governing miR expression within the context of OC and the current status of epigenetic advancement in the therapeutic development and clinical trial progression. Through network analysis we elucidate the regulatory interactions between dysregulated miRs in OC and their targets which are involved in different signaling pathways. By exploring these interconnected facets and critical analysis, we endeavor to provide a nuanced understanding of the molecular dynamics underlying OC, its detection and shedding light on potential avenues for miRs and epigenetics-based therapeutic intervention and management strategies.
Collapse
Affiliation(s)
- Arpan Dey Bhowmik
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Pallab Shaw
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Mohan Shankar Gopinatha Pillai
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Geeta Rao
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Shailendra Kumar Dhar Dwivedi
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
6
|
Meng X, Liang X, Yang S, Wu D, Wang X. A miRNA-7704/IL2RB/AKT feedback loop regulates tumorigenesis and chemoresistance in ovarian cancer. Exp Cell Res 2024; 437:114012. [PMID: 38565343 DOI: 10.1016/j.yexcr.2024.114012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 02/08/2024] [Accepted: 03/22/2024] [Indexed: 04/04/2024]
Abstract
Ovarian cancer is one of the most common gynecological tumors worldwide. Despite the availability of multiple treatments for ovarian cancer, its resistance to chemotherapy remains a significant challenge. miRNAs play crucial roles in the initiation and progression of cancer by affecting processes such as differentiation, proliferation, and chemoresistance. According to microarray and qPCR analyses, miR-7704 is significantly downregulated in cisplatin-resistant cells compared to parental cells. In this study, we found that miR-7704 inhibited the proliferation and promoted cisplatin sensitivity of ovarian cancer cells in vitro and in vivo. Moreover, ectopic expression of miR-7704 had the same effect as IL2RB knockdown. Further mechanistic studies revealed that miR-7704 played an inhibitory role by regulating IL2RB expression to inactivate the AKT signaling pathway. Furthermore, IL2RB reversed the miR-7704 mediated resistance to cisplatin in ovarian cancer. Based on these findings, miR-7704 and IL2RB show the potential as novel therapeutic targets for ovarian cancer.
Collapse
Affiliation(s)
- Xuan Meng
- Phase I Clinical Trial Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China; Department of Oncology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Xiaoqing Liang
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Shengjie Yang
- Phase I Clinical Trial Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Dongsheng Wu
- Department of Emergency, Yantaishan Hospital, Yantai, Shandong, China
| | - Xinghe Wang
- Phase I Clinical Trial Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
7
|
Wang L, Wang X, Zhu X, Zhong L, Jiang Q, Wang Y, Tang Q, Li Q, Zhang C, Wang H, Zou D. Drug resistance in ovarian cancer: from mechanism to clinical trial. Mol Cancer 2024; 23:66. [PMID: 38539161 PMCID: PMC10976737 DOI: 10.1186/s12943-024-01967-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 02/22/2024] [Indexed: 04/05/2024] Open
Abstract
Ovarian cancer is the leading cause of gynecological cancer-related death. Drug resistance is the bottleneck in ovarian cancer treatment. The increasing use of novel drugs in clinical practice poses challenges for the treatment of drug-resistant ovarian cancer. Continuing to classify drug resistance according to drug type without understanding the underlying mechanisms is unsuitable for current clinical practice. We reviewed the literature regarding various drug resistance mechanisms in ovarian cancer and found that the main resistance mechanisms are as follows: abnormalities in transmembrane transport, alterations in DNA damage repair, dysregulation of cancer-associated signaling pathways, and epigenetic modifications. DNA methylation, histone modifications and noncoding RNA activity, three key classes of epigenetic modifications, constitute pivotal mechanisms of drug resistance. One drug can have multiple resistance mechanisms. Moreover, common chemotherapies and targeted drugs may have cross (overlapping) resistance mechanisms. MicroRNAs (miRNAs) can interfere with and thus regulate the abovementioned pathways. A subclass of miRNAs, "epi-miRNAs", can modulate epigenetic regulators to impact therapeutic responses. Thus, we also reviewed the regulatory influence of miRNAs on resistance mechanisms. Moreover, we summarized recent phase I/II clinical trials of novel drugs for ovarian cancer based on the abovementioned resistance mechanisms. A multitude of new therapies are under evaluation, and the preliminary results are encouraging. This review provides new insight into the classification of drug resistance mechanisms in ovarian cancer and may facilitate in the successful treatment of resistant ovarian cancer.
Collapse
Affiliation(s)
- Ling Wang
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
- Chongqing Specialized Medical Research Center of Ovarian Cancer, Chongqing, China
- Organoid Transformational Research Center, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Xin Wang
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
- Chongqing Specialized Medical Research Center of Ovarian Cancer, Chongqing, China
- Organoid Transformational Research Center, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Xueping Zhu
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
- Chongqing Specialized Medical Research Center of Ovarian Cancer, Chongqing, China
- Organoid Transformational Research Center, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Lin Zhong
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
- Chongqing Specialized Medical Research Center of Ovarian Cancer, Chongqing, China
- Organoid Transformational Research Center, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Qingxiu Jiang
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
- Chongqing Specialized Medical Research Center of Ovarian Cancer, Chongqing, China
- Organoid Transformational Research Center, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Ya Wang
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
- Chongqing Specialized Medical Research Center of Ovarian Cancer, Chongqing, China
- Organoid Transformational Research Center, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Qin Tang
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
- Chongqing Specialized Medical Research Center of Ovarian Cancer, Chongqing, China
- Organoid Transformational Research Center, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Qiaoling Li
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
- Chongqing Specialized Medical Research Center of Ovarian Cancer, Chongqing, China
- Organoid Transformational Research Center, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Cong Zhang
- Chongqing Specialized Medical Research Center of Ovarian Cancer, Chongqing, China
- Organoid Transformational Research Center, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
- Biological and Pharmaceutical Engineering, School of Medicine, Chongqing University, Chongqing, China
| | - Haixia Wang
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China.
- Chongqing Specialized Medical Research Center of Ovarian Cancer, Chongqing, China.
- Organoid Transformational Research Center, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China.
| | - Dongling Zou
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China.
- Chongqing Specialized Medical Research Center of Ovarian Cancer, Chongqing, China.
- Organoid Transformational Research Center, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China.
| |
Collapse
|
8
|
Alam S, Giri PK. Novel players in the development of chemoresistance in ovarian cancer: ovarian cancer stem cells, non-coding RNA and nuclear receptors. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:6. [PMID: 38434767 PMCID: PMC10905178 DOI: 10.20517/cdr.2023.152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 02/03/2024] [Accepted: 02/22/2024] [Indexed: 03/05/2024]
Abstract
Ovarian cancer (OC) ranks as the fifth leading factor for female mortality globally, with a substantial burden of new cases and mortality recorded annually. Survival rates vary significantly based on the stage of diagnosis, with advanced stages posing significant challenges to treatment. OC is primarily categorized as epithelial, constituting approximately 90% of cases, and correct staging is essential for tailored treatment. The debulking followed by chemotherapy is the prevailing treatment, involving platinum-based drugs in combination with taxanes. However, the efficacy of chemotherapy is hindered by the development of chemoresistance, both acquired during treatment (acquired chemoresistance) and intrinsic to the patient (intrinsic chemoresistance). The emergence of chemoresistance leads to increased mortality rates, with many advanced patients experiencing disease relapse shortly after initial treatment. This review delves into the multifactorial nature of chemoresistance in OC, addressing mechanisms involving transport systems, apoptosis, DNA repair, and ovarian cancer stem cells (OCSCs). While previous research has identified genes associated with these mechanisms, the regulatory roles of non-coding RNA (ncRNA) and nuclear receptors in modulating gene expression to confer chemoresistance have remained poorly understood and underexplored. This comprehensive review aims to shed light on the genes linked to different chemoresistance mechanisms in OC and their intricate regulation by ncRNA and nuclear receptors. Specifically, we examine how these molecular players influence the chemoresistance mechanism. By exploring the interplay between these factors and gene expression regulation, this review seeks to provide a comprehensive mechanism driving chemoresistance in OC.
Collapse
Affiliation(s)
| | - Pankaj Kumar Giri
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi 110068, India
| |
Collapse
|
9
|
Liu Y, Liu G. Targeting NEAT1 Affects the Sensitivity to PARPi in Serous Ovarian Cancer by Regulating the Homologous Recombination Repair Pathway. J Cancer 2024; 15:1397-1413. [PMID: 38356722 PMCID: PMC10861825 DOI: 10.7150/jca.91896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 12/16/2023] [Indexed: 02/16/2024] Open
Abstract
Objective: Patients initially sensitive to PARPi (PARP inhibitor) regain resistance because of homologous recombination (HR) restoration, although PARPi has a synthetic lethality effect on serous ovarian cancer cells with BRCA1/2 mutations. This study aimed to investigate the role of NEAT1 in HR function and whether targeting NEAT1 in serous ovarian cancer cells could increase PARPi sensitivity. Methods: Ovarian cancer patients' clinical information and the expression of NEAT1 were collected from The Cancer Genome Atlas (TCGA) and the International Cancer Genome Consortium (ICGC). Ovarian cancer (OC) cells HeyA8 and SKOV3 were silenced by transfecting NEAT1 ASO. QRT-PCR confirmed the mRNA expression of RAD51, FOXM1, NEAT1_1 and NEAT1_2. We assessed the expression of RAD51, FOXM1, and γ-H2AX by Western blotting and Immunofluorescence. Comet Assays were used to detect DNA double-strand damage levels. In OC cells transfected with NEAT1 ASO or co-transfected overexpression RAD51/empty vector and si-NEAT1/si-ctrl, the sensitivity to Olaparib was determined using CCK8 assay. The Kaplan-Meier survival curves assessed the prognostic and predictive roles of NEAT1 in OC. Results: NEAT1 was an independent prognostic marker of ovarian cancer. NEAT1 knockdown reduced the expression of NEAT1_1, NEAT1_2, RAD51, and FOXM1 and increased the expression of γ-H2AX. In addition, Olaparib increased the expression of RAD51, representing HR repair efficiency, which was inhibited by NEAT1 knockdown. Moreover, the knockdown of NEAT1 increased the DNA damage caused by Olaparib, demonstrated by increased nuclear γ-H2AX foci, DNA in the tail, and expression of γ-H2AX. NEAT1 knockdown sensitized ovarian cancer cells to Olaparib by targeting RAD51-HR. NEAT1 expression could predict response to chemotherapy for ovarian cancer. Conclusions: NEAT1 knockdown inhibited HR capacity and increased DNA damage caused by Olaparib in serous ovarian cancer cells, making them more sensitive to Olaparib and providing a crucial therapeutic advantage of increasing sensitivity to Olaparib.
Collapse
Affiliation(s)
- Yang Liu
- Departments of Obstetrics and Gynecology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Guoyan Liu
- Correspondence to: Dr. Guoyan Liu, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Department of Gynecologic Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Huanhuxi Road, Hexi District, Tianjin, 300060, China
| |
Collapse
|
10
|
Alshahrani SH, Yuliastanti T, Al-Dolaimy F, Korotkova NL, Rasulova I, Almuala AF, Alsaalamy A, Ali SHJ, Alasheqi MQ, Mustafa YF. A glimpse into let-7e roles in human disorders; friend or foe? Pathol Res Pract 2024; 253:154992. [PMID: 38103367 DOI: 10.1016/j.prp.2023.154992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/17/2023] [Accepted: 11/27/2023] [Indexed: 12/19/2023]
Abstract
MicroRNAs (miRNAs) have been linked to abnormal expression and regulation in a number of diseases, including cancer. Recent studies have concentrated on miRNA Let-7e's significance in precision medicine for cancer screening and diagnosis as well as its prognostic and therapeutic potential. Differential let-7e levels in bodily fluids have the possibility to enable early detection of cancer utilizing less-invasive techniques, reducing biopsy-related risks. Although Let-7e miRNAs have been described as tumor suppressors, it is crucial to note that there exists proof to support their oncogenic activity in vitro and in in vivo. Let-7e's significance in chemo- and radiation treatment decisions has also been demonstrated. Let-7e can also prevent the synthesis of proinflammatory cytokines in a number of degenerative disorders, including musculoskeletal and neurological conditions. For the first time, an overview of the significance of let-7e in the prevention, detection, and therapy of cancer and other conditions has been given in the current review. Additionally, we focused on the specific molecular processes that underlie the actions of let-7e, more particularly, on malignant cells.
Collapse
Affiliation(s)
| | | | | | - Nadezhda L Korotkova
- I.M. Sechenov First Moscow State Medical University, Moscow, Russian Federation; Federal State Budgetary Educational Institution of Higher Education "Privolzhsky Research Medical University" of the Ministry of Health of the Russian Federation, Nizhny Novgorod, Russian Federation
| | - Irodakhon Rasulova
- School of Humanities, Natural & Social Sciences, New Uzbekistan University, 54 Mustaqillik Ave., Tashkent 100007, Uzbekistan; Department of Public Health, Samarkand State Medical University, Amir Temur Street 18, Samarkand, Uzbekistan
| | - Abbas Firras Almuala
- College of Technical Engineering, the Islamic University, Najaf, Iraq; College of Technical Engineering, the Islamic University of Al Diwaniyah, Iraq; College of Technical Engineering, the Islamic University of Babylon, Iraq
| | - Ali Alsaalamy
- College of Technical Engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna 66002, Iraq
| | - Saad Hayif Jasim Ali
- Department of Medical Laboratory, College of Health and Medical Technololgy, Al-Ayen University, Thi-Qar, Iraq
| | | | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul 41001, Iraq
| |
Collapse
|
11
|
Zhang X, Ma D, Xuan B, Shi D, He J, Yu M, Xiong H, Ma Y, Shen C, Guo F, Cao Y, Yan Y, Gao Z, Tong T, Zhu X, Fang JY, Chen H, Hong J. LncRNA CACClnc promotes chemoresistance of colorectal cancer by modulating alternative splicing of RAD51. Oncogene 2023; 42:1374-1391. [PMID: 36906654 DOI: 10.1038/s41388-023-02657-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 02/24/2023] [Accepted: 02/27/2023] [Indexed: 03/13/2023]
Abstract
Long non-coding RNAs (lncRNAs) play important roles in carcinogenesis. However, the effect of lncRNA on chemoresistance and RNA alternative splicing remains largely unknown. In this study, we identified a novel lncRNA, CACClnc, which was upregulated and associated with chemoresistance and poor prognosis in colorectal cancer (CRC). CACClnc promoted CRC resistance to chemotherapy via promoting DNA repair and enhancing homologous recombination in vitro and in vivo. Mechanistically, CACClnc specifically bound to Y-box binding protein 1 (YB1, a splicing factor) and U2AF65 (a subunit of U2AF splicing factor), promoting the interaction between YB1 and U2AF65, and then modulated alternative splicing (AS) of RAD51 mRNA, and consequently altered CRC cell biology. In addition, expression of exosomal CACClnc in peripheral plasma of CRC patients can effectively predict the chemotherapy effect of patients before treatment. Thus, measuring and targeting CACClnc and its associated pathway could yield valuable insight into clinical management and might ameliorate CRC patients' outcomes.
Collapse
Affiliation(s)
- Xinyu Zhang
- State Key Laboratory for Oncogenes and Related Genes, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Dan Ma
- State Key Laboratory for Oncogenes and Related Genes, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Department of Gastroenterology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Baoqin Xuan
- State Key Laboratory for Oncogenes and Related Genes, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Debing Shi
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jie He
- Guangzhou Key Laboratory of Digestive Disease, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital and The Second Affiliated Hospital, South China University of Technology School of Medicine, Guangzhou, China
| | - Minhao Yu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hua Xiong
- State Key Laboratory for Oncogenes and Related Genes, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yanru Ma
- State Key Laboratory for Oncogenes and Related Genes, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chaoqin Shen
- State Key Laboratory for Oncogenes and Related Genes, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fangfang Guo
- State Key Laboratory for Oncogenes and Related Genes, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yingying Cao
- State Key Laboratory for Oncogenes and Related Genes, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuqing Yan
- State Key Laboratory for Oncogenes and Related Genes, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ziyun Gao
- State Key Laboratory for Oncogenes and Related Genes, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tianying Tong
- State Key Laboratory for Oncogenes and Related Genes, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoqiang Zhu
- State Key Laboratory for Oncogenes and Related Genes, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing-Yuan Fang
- State Key Laboratory for Oncogenes and Related Genes, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Haoyan Chen
- State Key Laboratory for Oncogenes and Related Genes, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Jie Hong
- State Key Laboratory for Oncogenes and Related Genes, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
12
|
Nasrpour Navaei Z, Taghehchian N, Zangouei AS, Abbaszadegan MR, Moghbeli M. MicroRNA-506 as a tumor suppressor in anaplastic thyroid carcinoma by regulation of WNT and NOTCH signaling pathways. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:594-602. [PMID: 37051101 PMCID: PMC10083834 DOI: 10.22038/ijbms.2023.69174.15069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 02/12/2023] [Indexed: 04/14/2023]
Abstract
OBJECTIVES Anaplastic thyroid carcinoma (ATC) is an aggressive thyroid tumor type that has a poor prognosis due to its high therapeutic resistance. Since ATC accounts for half of thyroid cancer-related deaths, it is required to introduce novel therapeutic targets to increase survival in ATC patients. WNT and NOTCH signaling pathways are the pivotal regulators of cell proliferation and migration that can be regulated by microRNAs. We assessed the role of miR-506 in the regulation of cell migration, apoptosis, and drug resistance via NOTCH and WNT pathways in ATC cells. MATERIALS AND METHODS The levels of miR-506 expressions were assessed in ATC cells and tissues. The levels of NOTCH, WNT, and EMT-related gene expressions were also assessed in miR-506 ectopic expressed cells compared with controls. Cell migration and drug resistance were also evaluated to assess the role of miR-506 in the regulation of ATC aggressiveness. RESULTS There were significant miR-506 down-regulations in ATC cells and clinical samples compared with normal cells and margins. MiR-506 suppressed NOTCH and WNT signaling pathways through LEF1, DVL, FZD1, HEY2, HES5, and HEY2 down-regulations, and APC and GSK3b up-regulations. MiR-506 significantly inhibited ATC cell migration and EMT (P=0.028). Moreover, miR-506 significantly increased Cisplatin (P=0.004), Paclitaxel (P<0.0001), and Doxorubicin (P=0.0014) sensitivities in ATC cells. CONCLUSION MiR-506 regulated EMT, cell migration, and chemoresistance through regulation of WNT and NOTCH signaling pathways in ATC cells. Therefore, after confirmation with animal studies, it can be introduced as an efficient novel therapeutic factor for ATC tumors.
Collapse
Affiliation(s)
- Zahra Nasrpour Navaei
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negin Taghehchian
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Sadra Zangouei
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
13
|
Tavakoli Pirzaman A, Ebrahimzadeh Pirshahid M, Babajani B, Rahmati A, Niknezhad S, Hosseinzadeh R, Taheri M, Ebrahimi-Zadeh F, Doostmohamadian S, Kazemi S. The Role of microRNAs in Regulating Cancer Cell Response to Oxaliplatin-Containing Regimens. Technol Cancer Res Treat 2023; 22:15330338231206003. [PMID: 37849311 PMCID: PMC10586010 DOI: 10.1177/15330338231206003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/18/2023] [Accepted: 10/18/2023] [Indexed: 10/19/2023] Open
Abstract
Oxaliplatin (cyclohexane-1,2-diamine; oxalate; platinum [2+]) is a third-generation chemotherapeutic drug with anticancer effects. Oxaliplatin has a role in the treatment of several cancers. It is one of the few drugs which can eliminate the neoplastic cells of colorectal cancer. Also, it has an influential role in breast cancer, lung cancer, bladder cancer, prostate cancer, and gastric cancer. Although oxaliplatin has many beneficial effects in cancer treatment, resistance to this drug is in the way to cure neoplastic cells and reduce treatment efficacy. microRNAs are a subtype of small noncoding RNAs with ∼22 nucleotides that exist among species. They have diverse roles in physiological processes, including cellular proliferation and cell death. Moreover, miRNAs have essential roles in resistance to cancer treatment and can strengthen sensitivity to chemotherapeutic drugs and regimens. In colorectal cancer, the co-treatment of oxaliplatin with anti-miR-19a can partially reverse the oxaliplatin resistance through the upregulation of phosphatase and tensin homolog (PTEN). Moreover, by preventing the spread of gastric cancer cells and downregulating glypican-3 (GPC3), MiR-4510 may modify immunosuppressive signals in the tumor microenvironment. Treatment with oxaliplatin may develop into a specialized therapeutic drug for patients with miR-4510 inhibition and glypican-3-expressing gastric cancer. Eventually, miR-122 upregulation or Wnt/β-catenin signaling suppression boosted the death of HCC cells and made them more sensitive to oxaliplatin. Herein, we have reviewed the role of microRNAs in regulating cancer cells' response to oxaliplatin, with particular attention to gastrointestinal cancers. We also discussed the role of these noncoding RNAs in the pathophysiology of oxaliplatin-induced neuropathic pain.
Collapse
Affiliation(s)
| | | | - Bahareh Babajani
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Amirhossein Rahmati
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Shokat Niknezhad
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Rezvan Hosseinzadeh
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Mehdi Taheri
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Faezeh Ebrahimi-Zadeh
- Student Research Committee, school of Medicine, Jahrom University of Medical Science, Jahrom, Iran
| | | | - Sohrab Kazemi
- Cellular and Molecular Biology Research Center, Health Research Center, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
14
|
Quercetin and Isorhamnetin Reduce Benzo[a]pyrene-Induced Genotoxicity by Inducing RAD51 Expression through Downregulation of miR-34a. Int J Mol Sci 2022; 23:ijms232113125. [PMID: 36361910 PMCID: PMC9653982 DOI: 10.3390/ijms232113125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/22/2022] [Accepted: 10/25/2022] [Indexed: 01/24/2023] Open
Abstract
Benzo[a]pyrene (B[a]P) is metabolized in the liver into highly reactive mutagenic and genotoxic metabolites, which induce carcinogenesis. The mutagenic factors, including B[a]P-7,8-dihydrodiol-9,10-epoxide (BPDE) and reactive oxygen species, generated during B[a]P metabolism can cause DNA damage, such as BPDE-DNA adducts, 8-oxo-dG, and double-strand breaks (DSBs). In this study, we mechanistically investigated the effects of quercetin and its major metabolite isorhamnetin on the repair of B[a]P-induced DNA DSBs. Whole-transcriptome analysis showed that quercetin and isorhamnetin each modulate the expression levels of genes involved in DNA repair, especially those in homologous recombination. RAD51 was identified as a key gene whose expression level was decreased in B[a]P-treated cells and increased by quercetin or isorhamnetin treatment. Furthermore, the number of γH2AX foci induced by B[a]P was significantly decreased by quercetin or isorhamnetin, whereas RAD51 mRNA and protein levels were increased. Additionally, among the five microRNAs (miRs) known to downregulate RAD51, miR-34a level was significantly downregulated by quercetin or isorhamnetin. The protective effect of quercetin or isorhamnetin was lower in cells transfected with a miR-34a mimic than in non-transfected cells, and the B[a]P-induced DNA DSBs remained unrepaired. Our results show that quercetin and isorhamnetin each upregulates RAD51 by downregulating miR-34a and thereby suppresses B[a]P-induced DNA damage.
Collapse
|
15
|
Fu Y, Liu H, Long M, Song L, Meng Z, Lin S, Zhang Y, Qin J. Icariin attenuates the tumor growth by targeting miR-1-3p/TNKS2/Wnt/β-catenin signaling axis in ovarian cancer. Front Oncol 2022; 12:940926. [PMID: 36185280 PMCID: PMC9516086 DOI: 10.3389/fonc.2022.940926] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/18/2022] [Indexed: 11/20/2022] Open
Abstract
Purpose Despite various therapy advances, ovarian cancer remains an incurable disease for which survival rates have only modestly improved. Natural products are important sources of anti-cancer lead compounds. Icariin exhibited broad anti-cancer efficacy. However, the mechanism of icariin against ovarian cancer is poorly elucidated. Methods Cell viability was detected to evaluate the effect of icariin on SKOV-3 cells. The cell cycle and apoptosis were analyzed. The transcript of SKOV-3 cells was profiled by RNA-seq. GSEA and DEGs analyses were performed to interpret gene expression data. Western blot and TOP/FOP flash assay were applied to detect Wnt/β-catenin signaling. MiRDB database and dual-luciferase reporter assay was applied to study the regulation of miR-1-3p on TNKS2. Anti-tumor efficacy of icariin was evaluated by xenograft mouse model. Immunohistochemistry was performed with antibodies against Ki67. Results Icariin significantly suppressed the proliferation of SKOV-3 cells. Furthermore, icariin stalled cell cycle and induced apoptosis by blocking TNKS2/Wnt/β-catenin pathway through upregulating the level of miR-1-3p. Finally, icariin dramatically suppressed tumor growth in vivo. Conclusions In this study, we demonstrated for the first time that icariin significantly attenuated the growth of ovarian tumor in xenograft mouse model. Furthermore, we systematically revealed that icariin attenuates the tumor progression by suppressing TNKS2/Wnt/β-catenin signaling via upregulating the level of miR-1-3p in ovarian cancer with transcriptome analysis.
Collapse
Affiliation(s)
- Yanjin Fu
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Haiquan Liu
- Huizhou Traditional Chinese Medicine Hospital, Guangzhou University of Traditional Chinese Medicine, Huizhou, Guangdong, China
| | - Mengsha Long
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Linliang Song
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Zuyu Meng
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Shaozi Lin
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Yiyao Zhang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - JiaJia Qin
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
- *Correspondence: JiaJia Qin,
| |
Collapse
|
16
|
Bagnoli M, Nicoletti R, Valitutti M, Rizzo A, Napoli A, Montalvão De Azevedo R, Tomassetti A, Mezzanzanica D. Impairment of RAD17 Functions by miR-506-3p as a Novel Synthetic Lethal Approach Targeting DNA Repair Pathways in Ovarian Cancer. Front Oncol 2022; 12:923508. [PMID: 35924161 PMCID: PMC9340372 DOI: 10.3389/fonc.2022.923508] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/23/2022] [Indexed: 11/13/2022] Open
Abstract
Epithelial ovarian cancer (EOC) remains the most lethal gynecological cancer and development of chemo-resistance is a major factor in disease relapse. Homologous recombination (HR) is a critical pathway for DNA double strand break repair and its deficiency is associated to a better response to DNA damage-inducing agents. Strategies to inhibit HR-mediated DNA repair is a clinical need to improve patients’ outcome. MicroRNA (miRNAs) affect most of cellular processes including response to cancer treatment. We previously showed that miR-506-3p targets RAD51, an essential HR component. In this study we demonstrated that: i) another HR component, RAD17, is also a direct target of miR-506-3p and that it is involved in mediating miR-506-3p phenotypic effects; ii) the impairment of miR-506-3p binding to RAD17 3’ UTR reverted the miR-506-3p induced platinum sensitization; iii) miR-506-3p/RAD17 axis reduces the ability of EOC cell to sense DNA damage, abrogates the G2/M cell cycle checkpoint thus delaying the G2/M cell cycle arrest likely allowing the entry into mitosis of heavily DNA-damaged cells with a consequent mitotic catastrophe; iv) RAD17 expression, regulated by miR-506-3p, is synthetically lethal with inhibitors of cell cycle checkpoint kinases Chk1 and Wee1 in platinum resistant cell line. Overall miR-506-3p expression may recapitulate a BRCAness phenotype sensitizing EOC cells to chemotherapy and helping in selecting patients susceptible to DNA damaging drugs in combination with new small molecules targeting DNA-damage repair pathway.
Collapse
|
17
|
Huang Z, Zhang Z, Zhou C, Liu L, Huang C. Epithelial–mesenchymal transition: The history, regulatory mechanism, and cancer therapeutic opportunities. MedComm (Beijing) 2022; 3:e144. [PMID: 35601657 PMCID: PMC9115588 DOI: 10.1002/mco2.144] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 02/05/2023] Open
Abstract
Epithelial–mesenchymal transition (EMT) is a program wherein epithelial cells lose their junctions and polarity while acquiring mesenchymal properties and invasive ability. Originally defined as an embryogenesis event, EMT has been recognized as a crucial process in tumor progression. During EMT, cell–cell junctions and cell–matrix attachments are disrupted, and the cytoskeleton is remodeled to enhance mobility of cells. This transition of phenotype is largely driven by a group of key transcription factors, typically Snail, Twist, and ZEB, through epigenetic repression of epithelial markers, transcriptional activation of matrix metalloproteinases, and reorganization of cytoskeleton. Mechanistically, EMT is orchestrated by multiple pathways, especially those involved in embryogenesis such as TGFβ, Wnt, Hedgehog, and Hippo, suggesting EMT as an intrinsic link between embryonic development and cancer progression. In addition, redox signaling has also emerged as critical EMT modulator. EMT confers cancer cells with increased metastatic potential and drug resistant capacity, which accounts for tumor recurrence in most clinic cases. Thus, targeting EMT can be a therapeutic option providing a chance of cure for cancer patients. Here, we introduce a brief history of EMT and summarize recent advances in understanding EMT mechanisms, as well as highlighting the therapeutic opportunities by targeting EMT in cancer treatment.
Collapse
Affiliation(s)
- Zhao Huang
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu 610041 China
| | - Zhe Zhang
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu 610041 China
| | - Chengwei Zhou
- Department of Thoracic Surgery the Affiliated Hospital of Medical School of Ningbo University Ningbo China
| | - Lin Liu
- Department of Thoracic Surgery the Affiliated Hospital of Medical School of Ningbo University Ningbo China
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu 610041 China
| |
Collapse
|
18
|
Interactions between miRNAs and Double-Strand Breaks DNA Repair Genes, Pursuing a Fine-Tuning of Repair. Int J Mol Sci 2022; 23:ijms23063231. [PMID: 35328651 PMCID: PMC8954595 DOI: 10.3390/ijms23063231] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/06/2022] [Accepted: 03/09/2022] [Indexed: 02/04/2023] Open
Abstract
The repair of DNA damage is a crucial process for the correct maintenance of genetic information, thus, allowing the proper functioning of cells. Among the different types of lesions occurring in DNA, double-strand breaks (DSBs) are considered the most harmful type of lesion, which can result in significant loss of genetic information, leading to diseases, such as cancer. DSB repair occurs through two main mechanisms, called non-homologous end joining (NHEJ) and homologous recombination repair (HRR). There is evidence showing that miRNAs play an important role in the regulation of genes acting in NHEJ and HRR mechanisms, either through direct complementary binding to mRNA targets, thus, repressing translation, or by targeting other genes involved in the transcription and activity of DSB repair genes. Therefore, alteration of miRNA expression has an impact on the ability of cells to repair DSBs, which, in turn, affects cancer therapy sensitivity. This latter gives account of the importance of miRNAs as regulators of NHEJ and HRR and places them as a promising target to improve cancer therapy. Here, we review recent reports demonstrating an association between miRNAs and genes involved in NHEJ and HRR. We employed the Web of Science search query TS (“gene official symbol/gene aliases*” AND “miRNA/microRNA/miR-”) and focused on articles published in the last decade, between 2010 and 2021. We also performed a data analysis to represent miRNA–mRNA validated interactions from TarBase v.8, in order to offer an updated overview about the role of miRNAs as regulators of DSB repair.
Collapse
|
19
|
Sun Y, Meng C, Liu G. MicroRNA-506-3p inhibits ovarian cancer metastasis by down-regulating the expression of EZH2. J Cancer 2022; 13:943-950. [PMID: 35154460 PMCID: PMC8824902 DOI: 10.7150/jca.66959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 12/11/2021] [Indexed: 11/26/2022] Open
Abstract
Objective: To investigate the role of miR-506-3p in ovarian cancer (OvCa) metastasis. Methods: We overexpressed miR-506-3p in OvCa cells, and cell migration and invasion capacities were assessed in vitro using Transwell assays and wound healing assay. EZH2 is a target of miR-506-3p. We overexpressed and knocked down EZH2 in SKOV3 cells, and assessed its impact on cell migration and invasion. The orthotopic OvCa mouse models were conducted to confirm the role of miR-506-3p in OvCa metastasis. Results: In this research, we found that miR-506-3p reduced EZH2 expression and obviously suppressed the cell migration and invasion in ovarian cancer (OvCa). Moreover, the knockout of EZH2 mimicked the effect of miR-506-3p on invasion and migration, whereas EZH2 overexpression rescued the inhibitory effect of miR-506-3p. The orthotopic OvCa mouse models and clinical cases also confirmed the negative correlation between miR-506-3p and EZH2 in OvCa Conclusions: MiR-506-3p can suppress cell migration and invasion by targeting EZH2 in OvCa. Our study provides evidence supporting miR-506-3p-based therapy in OvCa.
Collapse
Affiliation(s)
- Yue Sun
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin, 300052, China
| | - Chao Meng
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin, 300052, China
| | - Guoyan Liu
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin, 300052, China
| |
Collapse
|
20
|
Han Y, Hu X, Yun X, Liu J, Yang J, Tian Z, Zhang X, Zhang Y, Wang X. Nucleolar and spindle associated protein 1 enhances chemoresistance through DNA damage repair pathway in chronic lymphocytic leukemia by binding with RAD51. Cell Death Dis 2021; 12:1083. [PMID: 34782617 PMCID: PMC8593035 DOI: 10.1038/s41419-021-04368-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 10/21/2021] [Accepted: 10/27/2021] [Indexed: 12/14/2022]
Abstract
Nucleolar and spindle-associated protein 1 (NUSAP1) is an essential regulator of mitotic progression, spindle assembly, and chromosome attachment. Although NUSAP1 acts as an oncogene involved in the progression of several cancers, the exact role of chronic lymphocytic leukemia (CLL) remains elusive. Herein, we first discovered obvious overexpression of NUSAP1 in CLL associated with poor prognosis. Next, the NUSAP1 level was modulated by transfecting CLL cells with lentivirus. Silencing NUSAP1 inhibited the cell proliferation, promoted cell apoptosis and G0/G1 phase arrest. Mechanistically, high expression of NUSAP1 strengthened DNA damage repairing with RAD51 engagement. Our results also indicated that NUSAP1 knockdown suppressed the growth CLL cells in vivo. We further confirmed that NUSAP1 reduction enhanced the sensitivity of CLL cells to fludarabine or ibrutinib. Overall, our research investigates the mechanism by which NUSAP1 enhances chemoresistance via DNA damage repair (DDR) signaling by stabilizing RAD51 in CLL cells. Hence, NUSAP1 may be expected to be a perspective target for the treatment of CLL with chemotherapy resistance.
Collapse
Affiliation(s)
- Yang Han
- grid.460018.b0000 0004 1769 9639Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021 China ,grid.460018.b0000 0004 1769 9639Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021 China ,grid.27255.370000 0004 1761 1174School of Medicine, Shandong University, Jinan, Shandong 250012 China ,Shandong Provincial Engineering Research Center of Lymphoma, Jinan, Shandong 250021 China
| | - Xinting Hu
- grid.460018.b0000 0004 1769 9639Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021 China ,grid.460018.b0000 0004 1769 9639Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021 China ,grid.27255.370000 0004 1761 1174School of Medicine, Shandong University, Jinan, Shandong 250012 China ,Shandong Provincial Engineering Research Center of Lymphoma, Jinan, Shandong 250021 China
| | - Xiaoya Yun
- grid.460018.b0000 0004 1769 9639Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021 China ,grid.460018.b0000 0004 1769 9639Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021 China ,grid.27255.370000 0004 1761 1174School of Medicine, Shandong University, Jinan, Shandong 250012 China ,Shandong Provincial Engineering Research Center of Lymphoma, Jinan, Shandong 250021 China
| | - Jiarui Liu
- grid.460018.b0000 0004 1769 9639Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021 China ,grid.460018.b0000 0004 1769 9639Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021 China ,grid.27255.370000 0004 1761 1174School of Medicine, Shandong University, Jinan, Shandong 250012 China ,Shandong Provincial Engineering Research Center of Lymphoma, Jinan, Shandong 250021 China
| | - Juan Yang
- grid.460018.b0000 0004 1769 9639Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021 China ,grid.460018.b0000 0004 1769 9639Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021 China ,grid.27255.370000 0004 1761 1174School of Medicine, Shandong University, Jinan, Shandong 250012 China ,Shandong Provincial Engineering Research Center of Lymphoma, Jinan, Shandong 250021 China
| | - Zheng Tian
- grid.460018.b0000 0004 1769 9639Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021 China ,grid.460018.b0000 0004 1769 9639Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021 China ,grid.27255.370000 0004 1761 1174School of Medicine, Shandong University, Jinan, Shandong 250012 China ,Shandong Provincial Engineering Research Center of Lymphoma, Jinan, Shandong 250021 China
| | - Xin Zhang
- grid.460018.b0000 0004 1769 9639Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021 China ,grid.460018.b0000 0004 1769 9639Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021 China ,grid.27255.370000 0004 1761 1174School of Medicine, Shandong University, Jinan, Shandong 250012 China ,Shandong Provincial Engineering Research Center of Lymphoma, Jinan, Shandong 250021 China
| | - Ya Zhang
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China. .,Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China. .,School of Medicine, Shandong University, Jinan, Shandong, 250012, China. .,Shandong Provincial Engineering Research Center of Lymphoma, Jinan, Shandong, 250021, China. .,Branch of National Clinical Research Center for Hematologic Diseases, Jinan, Shandong, 250021, China. .,National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China. .,Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China. .,School of Medicine, Shandong University, Jinan, Shandong, 250012, China. .,Shandong Provincial Engineering Research Center of Lymphoma, Jinan, Shandong, 250021, China. .,Branch of National Clinical Research Center for Hematologic Diseases, Jinan, Shandong, 250021, China. .,National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
| |
Collapse
|
21
|
Palleschi M, Tedaldi G, Sirico M, Virga A, Ulivi P, De Giorgi U. Moving beyond PARP Inhibition: Current State and Future Perspectives in Breast Cancer. Int J Mol Sci 2021; 22:ijms22157884. [PMID: 34360649 PMCID: PMC8346118 DOI: 10.3390/ijms22157884] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 12/27/2022] Open
Abstract
Breast cancer is the most frequent and lethal tumor in women and finding the best therapeutic strategy for each patient is an important challenge. PARP inhibitors (PARPis) are the first, clinically approved drugs designed to exploit synthetic lethality in tumors harboring BRCA1/2 mutations. Recent evidence indicates that PARPis have the potential to be used both in monotherapy and combination strategies in breast cancer treatment. In this review, we show the mechanism of action of PARPis and discuss the latest clinical applications in different breast cancer treatment settings, including the use as neoadjuvant and adjuvant approaches. Furthermore, as a class, PARPis show many similarities but also certain critical differences which can have essential clinical implications. Finally, we report the current knowledge about the resistance mechanisms to PARPis. A systematic PubMed search, using the entry terms “PARP inhibitors” and “breast cancer”, was performed to identify all published clinical trials (Phase I-II-III) and ongoing trials (ClinicalTrials.gov), that have been reported and discussed in this review.
Collapse
Affiliation(s)
- Michela Palleschi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.P.); (M.S.); (U.D.G.)
| | - Gianluca Tedaldi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (A.V.); (P.U.)
- Correspondence: ; Tel.: +39-0543-739232; Fax: +39-0543-739221
| | - Marianna Sirico
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.P.); (M.S.); (U.D.G.)
| | - Alessandra Virga
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (A.V.); (P.U.)
| | - Paola Ulivi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (A.V.); (P.U.)
| | - Ugo De Giorgi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.P.); (M.S.); (U.D.G.)
| |
Collapse
|
22
|
Xing Y, Ruan G, Ni H, Qin H, Chen S, Gu X, Shang J, Zhou Y, Tao X, Zheng L. Tumor Immune Microenvironment and Its Related miRNAs in Tumor Progression. Front Immunol 2021; 12:624725. [PMID: 34084160 PMCID: PMC8167795 DOI: 10.3389/fimmu.2021.624725] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 04/29/2021] [Indexed: 12/11/2022] Open
Abstract
MiRNA is a type of small non-coding RNA, by regulating downstream gene expression that affects the progression of multiple diseases, especially cancer. MiRNA can participate in the biological processes of tumor, including proliferation, invasion and escape, and exhibit tumor enhancement or inhibition. The tumor immune microenvironment contains numerous immune cells. These cells include lymphocytes with tumor suppressor effects such as CD8+ T cells and natural killer cells, as well as some tumor-promoting cells with immunosuppressive functions, such as regulatory T cells and myeloid-derived suppressor cells. MiRNA can affect the tumor immune microenvironment by regulating the function of immune cells, which in turn modulates the progression of tumor cells. Investigating the role of miRNA in regulating the tumor immune microenvironment will help elucidate the specific mechanisms of interaction between immune cells and tumor cells, and may facilitate the use of miRNA as a predictor of immune disorders in tumor progression. This review summarizes the multifarious roles of miRNA in tumor progression through regulation of the tumor immune microenvironment, and provides guidance for the development of miRNA drugs to treat tumors and for the use of miRNA as an auxiliary means in tumor immunotherapy.
Collapse
Affiliation(s)
- Yingying Xing
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Yoshida K, Yokoi A, Yamamoto Y, Kajiyama H. ChrXq27.3 miRNA cluster functions in cancer development. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:112. [PMID: 33766100 PMCID: PMC7992321 DOI: 10.1186/s13046-021-01910-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/14/2021] [Indexed: 12/11/2022]
Abstract
MicroRNAs (miRNAs) regulate the expression of their target genes post-transcriptionally; thus, they are deeply involved in fundamental biological processes. miRNA clusters contain two or more miRNA-encoding genes, and these miRNAs are usually coexpressed due to common expression mechanisms. Therefore, miRNA clusters are effective modulators of biological pathways by the members coordinately regulating their multiple target genes, and an miRNA cluster located on the X chromosome q27.3 region has received much attention in cancer research recently. In this review, we discuss the novel findings of the chrXq27.3 miRNA cluster in various types of cancer. The chrXq27.3 miRNA cluster contains 30 mature miRNAs synthesized from 22 miRNA-encoding genes in an ~ 1.3-Mb region. The expressions of these miRNAs are usually negligible in many normal tissues, with the male reproductive system being an exception. In cancer tissues, each miRNA is dysregulated, compared with in adjacent normal tissues. The miRNA-encoding genes are not uniformly distributed in the region, and they are further divided into two groups (the miR-506-514 and miR-888-892 groups) according to their location on the genome. Most of the miRNAs in the former group are tumor-suppressive miRNAs that are further downregulated in various cancers compared with normal tissues. miR-506-3p in particular is the most well-known miRNA in this cluster, and it has various tumor-suppressive functions associated with the epithelial–mesenchymal transition, proliferation, and drug resistance. Moreover, other miRNAs, such as miR-508-3p and miR-509-3p, have similar tumor-suppressive effects. Hence, the expression of these miRNAs is clinically favorable as prognostic factors in various cancers. However, the functions of the latter group are less understood. In the latter group, miR-888-5p displays oncogenic functions, whereas miR-892b is tumor suppressive. Therefore, the functions of the miR-888–892 group are considered to be cell type- or tissue-specific. In conclusion, the chrXq27.3 miRNA cluster is a critical regulator of cancer progression, and the miRNAs themselves, their regulatory mechanisms, and their target genes might be promising therapeutic targets.
Collapse
Affiliation(s)
- Kosuke Yoshida
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Tsuruma-cho 65, Showa-ku, Nagoya, 466-8550, Japan.,Institute for Advanced Research, Nagoya University, Nagoya, Japan.,Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Akira Yokoi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Tsuruma-cho 65, Showa-ku, Nagoya, 466-8550, Japan. .,Institute for Advanced Research, Nagoya University, Nagoya, Japan.
| | - Yusuke Yamamoto
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Tsuruma-cho 65, Showa-ku, Nagoya, 466-8550, Japan
| |
Collapse
|
24
|
Chu H, Han N, Xu J. CMPK1 Regulated by miR-130b Attenuates Response to 5-FU Treatment in Gastric Cancer. Front Oncol 2021; 11:637470. [PMID: 33816278 PMCID: PMC8013733 DOI: 10.3389/fonc.2021.637470] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 01/26/2021] [Indexed: 12/12/2022] Open
Abstract
Gastric cancer (GC) remains a major world-wide challenge, especially in Asian countries. Chemotherapy with 5-fluorouracil (5-FU) and cisplatin is used as the first-line treatment and development of chemoresistance is a major cause of progression. UMP/CMP kinase is responsible for the phosphorylation of the ribonucleotide metabolite 5-fluoro-5′-monophosphate (FUMP) in 5-FU metabolic process, and recognized as a key step in the conversion of 5-FU to cytotoxic metabolites. Our bioinformatics analysis and molecular experiments demonstrated that high expression of CMPK1 was associated with prolonged survival and response to 5-FU treatment in GC samples. Further analysis demonstrated that miR-130b as a key epigenetic regulator of CMPK1, and miR-130b-mediated attenuation of CMPK1 resulted in resistance of gastric cancer cells to DNA damage and cell death after treatment with 5-FU. Rescue experiments with augmented CMPK1 expression abolished the effect of miR-130b demonstrating the key function of this miRNA in this pathway. Thus, this newly identified miR-130b-CMPK1 axis suggests a potentially new chemotherapeutic strategy for improved response to 5-FU therapy.
Collapse
Affiliation(s)
- Huaizhu Chu
- Department of Oncological Surgery, Qinghai Provincial People's Hospital, Xining, China
| | - Nahui Han
- Department of Pain Medicine, Qinghai Provincial People's Hospital, Xining, China
| | - Jianguo Xu
- Department of Oncological Surgery, Qinghai Provincial People's Hospital, Xining, China
| |
Collapse
|
25
|
Gonzalez Bosquet J, Devor EJ, Newtson AM, Smith BJ, Bender DP, Goodheart MJ, McDonald ME, Braun TA, Thiel KW, Leslie KK. Creation and validation of models to predict response to primary treatment in serous ovarian cancer. Sci Rep 2021; 11:5957. [PMID: 33727600 PMCID: PMC7971042 DOI: 10.1038/s41598-021-85256-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 02/24/2021] [Indexed: 01/31/2023] Open
Abstract
Nearly a third of patients with high-grade serous ovarian cancer (HGSC) do not respond to initial therapy and have an overall poor prognosis. However, there are no validated tools that accurately predict which patients will not respond. Our objective is to create and validate accurate models of prediction for treatment response in HGSC. This is a retrospective case–control study that integrates comprehensive clinical and genomic data from 88 patients with HGSC from a single institution. Responders were those patients with a progression-free survival of at least 6 months after treatment. Only patients with complete clinical information and frozen specimen at surgery were included. Gene, miRNA, exon, and long non-coding RNA (lncRNA) expression, gene copy number, genomic variation, and fusion-gene determination were extracted from RNA-sequencing data. DNA methylation analysis was performed. Initial selection of informative variables was performed with univariate ANOVA with cross-validation. Significant variables (p < 0.05) were included in multivariate lasso regression prediction models. Initial models included only one variable. Variables were then combined to create complex models. Model performance was measured with area under the curve (AUC). Validation of all models was performed using TCGA HGSC database. By integrating clinical and genomic variables, we achieved prediction performances of over 95% in AUC. Most performances in the validation set did not differ from the training set. Models with DNA methylation or lncRNA underperformed in the validation set. Integrating comprehensive clinical and genomic data from patients with HGSC results in accurate and robust prediction models of treatment response.
Collapse
Affiliation(s)
- Jesus Gonzalez Bosquet
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Iowa Hospitals and Clinics, Iowa City, IA, 52242, USA. .,Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA, 52242, USA.
| | - Eric J Devor
- Department of Obstetrics and Gynecology, University of Iowa Hospitals and Clinics, Iowa City, IA, 52242, USA
| | - Andreea M Newtson
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Iowa Hospitals and Clinics, Iowa City, IA, 52242, USA
| | - Brian J Smith
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA, 52242, USA.,Department of Biostatistics, University of Iowa College of Public Health, Iowa City, IA, 52242, USA
| | - David P Bender
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Iowa Hospitals and Clinics, Iowa City, IA, 52242, USA.,Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA, 52242, USA
| | - Michael J Goodheart
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Iowa Hospitals and Clinics, Iowa City, IA, 52242, USA.,Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA, 52242, USA
| | - Megan E McDonald
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Iowa Hospitals and Clinics, Iowa City, IA, 52242, USA
| | - Terry A Braun
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA, 52242, USA.,Coordinated Laboratory for Computational Genomics, University of Iowa Hospitals and Clinics, Iowa City, IA, 52242, USA
| | - Kristina W Thiel
- Department of Obstetrics and Gynecology, University of Iowa Hospitals and Clinics, Iowa City, IA, 52242, USA
| | - Kimberly K Leslie
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA, 52242, USA.,Department of Obstetrics and Gynecology, University of Iowa Hospitals and Clinics, Iowa City, IA, 52242, USA
| |
Collapse
|
26
|
Zhang L, Wang M, Chen M, Niu W, Liu W, Leng T, Ji W, Lei B. A safe and efficient bioactive citrate-lysine/miRNA33 agonist nanosystem for high fat diet-induced obesity therapy. CHEMICAL ENGINEERING JOURNAL 2021; 408:127304. [DOI: 10.1016/j.cej.2020.127304] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2025]
|
27
|
Raue R, Frank AC, Syed SN, Brüne B. Therapeutic Targeting of MicroRNAs in the Tumor Microenvironment. Int J Mol Sci 2021; 22:ijms22042210. [PMID: 33672261 PMCID: PMC7926641 DOI: 10.3390/ijms22042210] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/16/2021] [Accepted: 02/18/2021] [Indexed: 02/06/2023] Open
Abstract
The tumor-microenvironment (TME) is an amalgamation of various factors derived from malignant cells and infiltrating host cells, including cells of the immune system. One of the important factors of the TME is microRNAs (miRs) that regulate target gene expression at a post transcriptional level. MiRs have been found to be dysregulated in tumor as well as in stromal cells and they emerged as important regulators of tumorigenesis. In fact, miRs regulate almost all hallmarks of cancer, thus making them attractive tools and targets for novel anti-tumoral treatment strategies. Tumor to stroma cell cross-propagation of miRs to regulate protumoral functions has been a salient feature of the TME. MiRs can either act as tumor suppressors or oncogenes (oncomiRs) and both miR mimics as well as miR inhibitors (antimiRs) have been used in preclinical trials to alter cancer and stromal cell phenotypes. Owing to their cascading ability to regulate upstream target genes and their chemical nature, which allows specific pharmacological targeting, miRs are attractive targets for anti-tumor therapy. In this review, we cover a recent update on our understanding of dysregulated miRs in the TME and provide an overview of how these miRs are involved in current cancer-therapeutic approaches from bench to bedside.
Collapse
Affiliation(s)
- Rebecca Raue
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (R.R.); (A.-C.F.)
| | - Ann-Christin Frank
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (R.R.); (A.-C.F.)
| | - Shahzad Nawaz Syed
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (R.R.); (A.-C.F.)
- Correspondence: (S.N.S.); (B.B.); Tel.: +49-69-6301-7424 (B.B.)
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (R.R.); (A.-C.F.)
- Project Group Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology, 60596 Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, 60590 Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe-University Frankfurt, 60596 Frankfurt, Germany
- Correspondence: (S.N.S.); (B.B.); Tel.: +49-69-6301-7424 (B.B.)
| |
Collapse
|
28
|
Sun Y, Wu J, Dong X, Zhang J, Meng C, Liu G. MicroRNA-506-3p increases the response to PARP inhibitors and cisplatin by targeting EZH2/β-catenin in serous ovarian cancers. Transl Oncol 2021; 14:100987. [PMID: 33360300 PMCID: PMC7770486 DOI: 10.1016/j.tranon.2020.100987] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 12/03/2020] [Accepted: 12/04/2020] [Indexed: 11/27/2022] Open
Abstract
Chemo-resistance is an important barrier to effective treatment of ovarian cancer. Poly (ADP-ribose) polymerase (PARP) inhibitors are currently promising targeted drugs used to treat BRCA-mutant ovarian cancer. Ovarian cancer patients with BRCA 1/2 mutations appear to benefit better from PARP inhibitors and chemotherapy. Understanding the mechanisms underlying PARP inhibitors and chemotherapy resistance is urgently needed. There is increasing evidence that microRNAs (miRNAs) are involved in drug resistance. MiR-506-3p is an effective inhibitor of the epithelial-to-mesenchymal transition (EMT), and can enhance chemotherapy and olaparib response in high-grade serous ovarian cancer (HGS-OvCa). Enhancer of Zeste Homolog 2 (EZH2) is considered as a direct target of miR-506-3p. The silencing of EZH2 mimics the inhibitory effects of miR-506-3p on chemo-resistance and olaparib response. Rescue of EZH2 prevented the functions of miR-506-3p. Moreover, EZH2 activates the β-catenin pathway. MiR-506-3p overexpression decreased the level of β-catenin, and the sensitivity to olaparib and cisplatin mediated by miR-506-3p was partially reversed by regulating β-catenin expression in ovarian cancer. Our results suggest that miR-506-3p increases response to PARP inhibitors and cisplatin in serous ovarian cancer by targeting EZH2/β-catenin signal pathway, which opens the possibility of using miR-506-3p overexpression as a potential therapeutic for ovarian cancer.
Collapse
Affiliation(s)
- Yue Sun
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jing Wu
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xiaoying Dong
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jingzi Zhang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Chao Meng
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Guoyan Liu
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, China.
| |
Collapse
|
29
|
Haque I, Kawsar HI, Motes H, Sharma M, Banerjee S, Banerjee SK, Godwin AK, Huang CH. Downregulation of miR-506-3p Facilitates EGFR-TKI Resistance through Induction of Sonic Hedgehog Signaling in Non-Small-Cell Lung Cancer Cell Lines. Int J Mol Sci 2020; 21:E9307. [PMID: 33291316 PMCID: PMC7729622 DOI: 10.3390/ijms21239307] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/04/2020] [Accepted: 12/04/2020] [Indexed: 02/08/2023] Open
Abstract
Non-small-cell lung cancer (NSCLC) patients with epidermal growth factor receptor (EGFR) mutation eventually develop resistance to EGFR-targeted tyrosine kinase inhibitors (TKIs). Treatment resistance remains the primary obstacle to the successful treatment of NSCLC. Although drug resistance mechanisms have been studied extensively in NSCLC, the regulation of these mechanisms has not been completely understood. Recently, increasing numbers of microRNAs (miRNAs) are implicated in EGFR-TKI resistance, indicating that miRNAs may serve as novel targets and may hold promise as predictive biomarkers for anti-EGFR therapy. MicroRNA-506 (miR-506) has been identified as a tumor suppressor in many cancers, including lung cancer; however, the role of miR-506 in lung cancer chemoresistance has not yet been addressed. Here we report that miR-506-3p expression was markedly reduced in erlotinib-resistant (ER) cells. We identified Sonic Hedgehog (SHH) as a novel target of miR-506-3p, aberrantly activated in ER cells. The ectopic overexpression of miR-506-3p in ER cells downregulates SHH signaling, increases E-cadherin expression, and inhibits the expression of vimentin, thus counteracting the epithelial-mesenchymal transition (EMT)-mediated chemoresistance. Our results advanced our understanding of the molecular mechanisms underlying EGFR-TKI resistance and indicated that the miR-506/SHH axis might represent a novel therapeutic target for future EGFR mutated lung cancer treatment.
Collapse
Affiliation(s)
- Inamul Haque
- Cancer Research Unit, Veterans Affairs Medical Center, Kansas City, MO 64128, USA
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Hameem I Kawsar
- Division of Medical Oncology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Hannah Motes
- Cancer Research Unit, Veterans Affairs Medical Center, Kansas City, MO 64128, USA
- Kirksville College of Osteopathic Medicine, Andrew Taylor Still University, Jefferson St, Kirksville, MO 63501, USA
| | - Mukut Sharma
- Research Service, Veterans Affairs Medical Center, Kansas City, MO 64128, USA
| | - Snigdha Banerjee
- Cancer Research Unit, Veterans Affairs Medical Center, Kansas City, MO 64128, USA
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Sushanta K Banerjee
- Cancer Research Unit, Veterans Affairs Medical Center, Kansas City, MO 64128, USA
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Chao H Huang
- Cancer Research Unit, Veterans Affairs Medical Center, Kansas City, MO 64128, USA
- Division of Medical Oncology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
30
|
Wang D, Zhang KF, Du G, Wang J, Zhao J. Berberine enhances the radiosensitivity of osteosarcoma by targeting Rad51 and epithelial-mesenchymal transition. J Cancer Res Ther 2020; 16:215-221. [PMID: 32474504 DOI: 10.4103/jcrt.jcrt_293_19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Objective Osteosarcoma is a malignant bone tumor and is generally treated with radiotherapy combined with radiosensitizers. The aim of the present study was to investigate the radiosensitization effects of berberine on osteosarcoma cells and the role of Rad51 in radiosensitivity by berberine. Materials and Methods Cells from the human osteosarcoma cell line MG-63 were exposed to γ-ray irradiation (0, 2, 4, 6, and 8 Gy) and berberine (20 μM). Radiosensitivity was evaluated by determining cell viability using an MTT assay. Flow cytometry was used to determine cell cycle and apoptosis. Real-time PCR and western blot were performed to analyze the mRNA and protein expressions of Rad51. The protein levels of E-cadherin and vimentin were also measured to evaluate the epithelial-mesenchymal transition (EMT) process. Tumor invasion was determined by the Boyden chamber assay. Results Berberine exacerbated the decline in viability of MG-63 cells exposed to γ-rays irradiation at various concentrations (25, 50, 75, and 100 μmol/L) and induced cell cycle arrest in the G2/M phase as well as apoptosis. The mRNA and protein expressions of Rad51 were significantly decreased by berberine in MG-63 cells. Inhibition of Rad51 by B02 enhanced the radiosensitivity of MG-63 cells. Berberine inhibited their invasive capability as well as increased E-cadherin and decreased vimentin protein levels; this indicated that berberine suppressed the EMT process in MG-63 cells exposed to γ-rays irradiation. Conclusion Berberine enhances the radiosensitivity of MG-63 osteosarcoma cells. Rad51 is a potential target of berberine in the radiosensitization of osteosarcoma.
Collapse
Affiliation(s)
- Dapeng Wang
- Research Centre for Regenerative Medicine, Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, 530022 Guangxi, Taian, Shandong, China
| | - Ke Fen Zhang
- Department of Pathology, Taishan Sanatorium, Taian, Shandong, China
| | - Gang Du
- Research Centre for Regenerative Medicine, Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, 530022 Guangxi, Taian, Shandong, China
| | - Jun Wang
- Department of Oncology, The Central Hospital of Taian, Taian, Shandong, China
| | - Jinmin Zhao
- Research Centre for Regenerative Medicine, Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, 530022 Guangxi, Taian, Shandong, China
| |
Collapse
|
31
|
Garofoli M, Volpicella M, Guida M, Porcelli L, Azzariti A. The Role of Non-Coding RNAs as Prognostic Factor, Predictor of Drug Response or Resistance and Pharmacological Targets, in the Cutaneous Squamous Cell Carcinoma. Cancers (Basel) 2020; 12:cancers12092552. [PMID: 32911687 PMCID: PMC7565940 DOI: 10.3390/cancers12092552] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/02/2020] [Accepted: 09/04/2020] [Indexed: 12/12/2022] Open
Abstract
Cutaneous squamous cell carcinoma (CSCC) is the most common keratinocyte-derived skin cancer in the Caucasian population. Exposure to UV radiations (UVRs) represents the main risk carcinogenesis, causing a considerable accumulation of DNA damage in epidermal keratinocytes with an uncontrolled hyperproliferation and tumor development. The limited and rarely durable response of CSCC to the current therapeutic options has led researchers to look for new therapeutic strategies. Recently, the multi-omics approaches have contributed to the identification and prediction of the key role of non-coding RNAs (ncRNAs), such as microRNAs (miRNAs), circularRNAs (circRNAs) and long non-coding RNAs (lncRNAs) in the regulation of several cellular processes in different tumor types, including CSCC. ncRNAs can modulate transcriptional and post-transcriptional events by interacting either with each other or with DNA and proteins, such as transcription factors and RNA-binding proteins. In this review, the implication of ncRNAs in tumorigenesis and their potential role as diagnostic biomarkers and therapeutic targets in human CSCC are reported.
Collapse
Affiliation(s)
- Marianna Garofoli
- Experimental Pharmacology Laboratory, IRCCS IstitutoTumori Giovanni Paolo II, 70124 Bari, Italy; (M.G.); (L.P.)
| | - Mariateresa Volpicella
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy;
| | - Michele Guida
- Rare Tumors and Melanoma Unit, IRCCS IstitutoTumori Giovanni Paolo II, 70124 Bari, Italy;
| | - Letizia Porcelli
- Experimental Pharmacology Laboratory, IRCCS IstitutoTumori Giovanni Paolo II, 70124 Bari, Italy; (M.G.); (L.P.)
| | - Amalia Azzariti
- Experimental Pharmacology Laboratory, IRCCS IstitutoTumori Giovanni Paolo II, 70124 Bari, Italy; (M.G.); (L.P.)
- Correspondence: ; Tel.: +39-080-555-5986
| |
Collapse
|
32
|
Yoshida K, Yokoi A, Kato T, Ochiya T, Yamamoto Y. The clinical impact of intra- and extracellular miRNAs in ovarian cancer. Cancer Sci 2020; 111:3435-3444. [PMID: 32750177 PMCID: PMC7541008 DOI: 10.1111/cas.14599] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/17/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022] Open
Abstract
Ovarian cancer is the most lethal gynecological cancer due to lack of early screening methods and acquired drug resistance. MicroRNAs (miRNAs) are effective post‐transcriptional regulators that are transferred by extracellular vesicles, such as exosomes. Numerous studies have revealed that miRNAs are differentially expressed in epithelial ovarian cancer and act either as oncogenes or tumor suppressor genes. Cancer cells secrete exosomes containing miRNAs, which exert various effects on the components of the tumor microenvironment, including cancer‐associated fibroblasts, macrophages, and adipocytes. Conversely, cancer cells also receive exosomes from these cells. As a result of cell‐to‐cell communication, epithelial ovarian cancer acquires a more aggressive phenotype and resistance to multiple drugs. In addition, some circulating miRNAs are protected from RNase degradation in the peripheral blood and can be potential non‐invasive biomarkers. In particular, the combination of several circulating miRNAs enhances the accuracy of cancer screening. Likewise, comprehensive analyses revealed specific miRNA signatures in non‐epithelial ovarian tumors and several miRNAs contributing to alterations of carcinogenic pathways. Overall, miRNAs play a crucial role in ovarian cancer progression. In this review, we discuss the emerging roles of intra‐ and extracellular miRNAs in ovarian cancers. In the near future, miRNAs will be practical biomarkers and computational deep learning will help in the clinical application of miRNAs. Moreover, miRNAs are potential therapeutic targets and agents, and there are ongoing clinical trials of miRNA replacement therapy. Therefore, accelerating research on miRNA might improve the prognosis of patients with ovarian cancer.
Collapse
Affiliation(s)
- Kosuke Yoshida
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Akira Yokoi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomoyasu Kato
- Department of Gynecology, National Cancer Center Hospital, Tokyo, Japan
| | - Takahiro Ochiya
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Yusuke Yamamoto
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| |
Collapse
|
33
|
Gong X, Li W, Dong L, Qu F. CircUBAP2 promotes SEMA6D expression to enhance the cisplatin resistance in osteosarcoma through sponging miR-506-3p by activating Wnt/β-catenin signaling pathway. J Mol Histol 2020. [PMID: 32472335 DOI: 10.1007/s10735-020-09883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The occurrence of chemo-resistance is an essential reason for the high morbidity of osteosarcoma (OS) patients. Circular RNAs (circRNAs) have been involved in the regulation of chemo-resistance in cancers. Semaphorins 6D (SEMA6D) is abnormally expressed in many cancers. However, the roles of circUBAP2 and SEMA6D in the chemo-resistance of OS are still unclear. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to detect the expression levels of circUBAP2, SEMA6D and microRNA-506-3p (miR-506-3p). The cisplatin resistance and proliferation of cells were evaluated by 3-(4, 5-dimethyl-2 thiazolyl)-2, 5-diphenyl-2-H-tetrazolium bromide assay. Western blot analysis was performed to measure the protein levels of Wnt/β-catenin signaling pathway biomarkers and SEMA6D. Also, the apoptosis, migration and invasion of cells were assessed by Flow cytometry and Transwell assays, respectively. Besides, Dual-luciferase reporter assay was used to verify the interaction between miR-506-3p and circUBAP2 or SEMA6D. We found that the expression levels of circUBAP2 and SEMA6D were increased in cisplatin-resistant OS tissues and cells. Knockdown of circUBAP2 inhibited the cisplatin resistance, silenced Wnt/β-catenin signaling pathway, hindered cell proliferation, migration and invasion, and promoted apoptosis in cisplatin-resistant OS cells, all of which could be reversed by overexpression of SEMA6D. MiR-506-3p could be sponged by circUBAP2 and could target SEMA6D. The suppression of miR-506-3p overexpression on the progression of OS cisplatin resistance could be reversed by SEMA6D overexpression, while miR-506-3p inhibitor also could invert the inhibitory effect of circUBAP2 silencing on the progression of OS cisplatin resistance. In conclusion, CircUBAP2 and SEMA6D played active roles in the progression of OS cisplatin resistance through miR-506-3p, which might provide some new ideas for studying the countermeasures of OS resistance.
Collapse
Affiliation(s)
| | | | - Lin Dong
- Department of Pharmacy, Yantai Affiliated Hospital of Binzhou Medical University, No. 717 Jinbu Avenue, Mouping District, Yantai, 264000, Shandong, China.
| | - Fangfei Qu
- Department of Special Inspection, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| |
Collapse
|
34
|
Vigneron N, Vernon M, Meryet-Figuière M, Lambert B, Briand M, Louis MH, Krieger S, Joly F, Lheureux S, Blanc-Fournier C, Gauduchon P, Poulain L, Denoyelle C. Predictive Relevance of Circulating miR-622 in Patients with Newly Diagnosed and Recurrent High-Grade Serous Ovarian Carcinoma. Clin Chem 2020; 66:352-362. [PMID: 32040573 DOI: 10.1093/clinchem/hvz013] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 10/21/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Identifying patients with high-grade serous ovarian cancer (HGSOC) who will respond to treatment remains a clinical challenge. We focused on miR-622, a miRNA involved in the homologous recombination repair (HRR) pathway, and we assessed its predictive value in serum prior to first-line chemotherapy and at relapse. METHODS Serum miR-622 expression was assessed in serum prior to first-line platinum-based chemotherapy in a prospective multicenter study (miRNA Serum Analysis, miRSA, NCT01391351) and a retrospective cohort (Biological Resource Center, BRC), and was also studied at relapse. Progression-free survival (PFS) and overall survival (OS) were used as primary and secondary endpoints prior to first-line chemotherapy and OS as a primary endpoint at relapse. RESULTS The group with high serum miR-622 expression was associated with a significantly lower PFS (15.4 versus 24.4 months; adjusted HR 2.11, 95% CI 1.2 3.8, P = 0.015) and OS (29.7 versus 40.6 months; adjusted HR 7.68, 95% CI 2.2-26.2, P = 0.0011) in the miRSA cohort. In the BRC cohort, a high expression of miR-622 was also associated with a significantly lower OS (22.8 versus 35.9 months; adjusted HR 1.98, 95% CI 1.1-3.6, P = 0.026). At relapse, high serum miR-622 was associated with a significantly lower OS (7.9 versus 20.6 months; adjusted HR 3.15, 95% CI 1.4-7.2, P = 0.0062). Serum miR-622 expression is a predictive independent biomarker of response to platinum-based chemotherapy for newly diagnosed and recurrent HGSOC. CONCLUSIONS These results may open new perspectives for HGSOC patient stratification and monitoring of resistance to platinum-based and poly(ADP-ribose)-polymerase-inhibitor-maintenance therapies, facilitating better and personalized treatment decisions.
Collapse
Affiliation(s)
- Nicolas Vigneron
- Normandie Université, UNICAEN, Inserm U1086 ANTICIPE, Biology and Innovative Therapeutics for Ovarian Cancer (BioTICLA), Caen, France.,UNICANCER, Cancer Center F. Baclesse, Caen, France
| | - Mégane Vernon
- Normandie Université, UNICAEN, Inserm U1086 ANTICIPE, Biology and Innovative Therapeutics for Ovarian Cancer (BioTICLA), Caen, France.,UNICANCER, Cancer Center F. Baclesse, Caen, France
| | - Matthieu Meryet-Figuière
- Normandie Université, UNICAEN, Inserm U1086 ANTICIPE, Biology and Innovative Therapeutics for Ovarian Cancer (BioTICLA), Caen, France.,UNICANCER, Cancer Center F. Baclesse, Caen, France
| | - Bernard Lambert
- Normandie Université, UNICAEN, Inserm U1086 ANTICIPE, Biology and Innovative Therapeutics for Ovarian Cancer (BioTICLA), Caen, France.,UNICANCER, Cancer Center F. Baclesse, Caen, France.,CNRS, Normandy Delegation, France
| | - Mélanie Briand
- Normandie Université, UNICAEN, Inserm U1086 ANTICIPE, Biology and Innovative Therapeutics for Ovarian Cancer (BioTICLA), Caen, France.,UNICANCER, Cancer Center F. Baclesse, Caen, France
| | - Marie-Hélène Louis
- Normandie Université, UNICAEN, Inserm U1086 ANTICIPE, Biology and Innovative Therapeutics for Ovarian Cancer (BioTICLA), Caen, France.,UNICANCER, Cancer Center F. Baclesse, Caen, France
| | - Sophie Krieger
- UNICANCER, Cancer Center F. Baclesse, Caen, France.,UNICANCER, Cancer Center F. Baclesse, Biopathology Department, Caen, France.,Normandie Université, UNIROUEN, Inserm U1245, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | - Florence Joly
- Normandie Université, UNICAEN, Inserm U1086 ANTICIPE, Biology and Innovative Therapeutics for Ovarian Cancer (BioTICLA), Caen, France.,UNICANCER, Cancer Center F. Baclesse, Caen, France.,UNICANCER, Cancer Center F. Baclesse, Medical Oncology Department and Clinical Research Unit, Caen, France
| | - Stéphanie Lheureux
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Center, Toronto, ON, Canada
| | - Cécile Blanc-Fournier
- UNICANCER, Cancer Center F. Baclesse, Caen, France.,UNICANCER, Cancer Center F. Baclesse, Biopathology Department, Caen, France
| | - Pascal Gauduchon
- Normandie Université, UNICAEN, Inserm U1086 ANTICIPE, Biology and Innovative Therapeutics for Ovarian Cancer (BioTICLA), Caen, France.,UNICANCER, Cancer Center F. Baclesse, Caen, France
| | - Laurent Poulain
- Normandie Université, UNICAEN, Inserm U1086 ANTICIPE, Biology and Innovative Therapeutics for Ovarian Cancer (BioTICLA), Caen, France.,UNICANCER, Cancer Center F. Baclesse, Caen, France
| | - Christophe Denoyelle
- Normandie Université, UNICAEN, Inserm U1086 ANTICIPE, Biology and Innovative Therapeutics for Ovarian Cancer (BioTICLA), Caen, France.,UNICANCER, Cancer Center F. Baclesse, Caen, France
| |
Collapse
|
35
|
Xu L, Wu T, Lu S, Hao X, Qin J, Wang J, Zhang X, Liu Q, Kong B, Gong Y, Liu Z, Shao C. Mitochondrial superoxide contributes to oxidative stress exacerbated by DNA damage response in RAD51-depleted ovarian cancer cells. Redox Biol 2020; 36:101604. [PMID: 32554304 PMCID: PMC7303666 DOI: 10.1016/j.redox.2020.101604] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 05/18/2020] [Accepted: 06/05/2020] [Indexed: 12/16/2022] Open
Abstract
Ovarian cancer is the most lethal gynecological malignancy. Abnormal homologous recombination repair, high level of reactive oxygen species (ROS) and upregulation of antioxidant genes are characteristic features of ovarian cancer. However, the molecular mechanisms governing the redox homeostasis in ovarian cancer cells remain to be fully elucidated. We here demonstrated a critical role of RAD51, a protein essential for homologous recombination, in the maintenance of redox homeostasis. We found that RAD51 is overexpressed in high grade serous ovarian cancer and is associated with poor prognosis. Depletion or inhibition of RAD51 results in G2/M arrest, increased production of reactive oxygen species and accumulation of oxidative DNA damage. Importantly, antioxidant N-acetylcysteine (NAC) significantly attenuated the induction of DNA damage and the perturbation of proliferation caused by RAD51 depletion. We further demonstrated that RAD51 inhibition or depletion led to elevated production of mitochondrial superoxide and increased accumulation of mitochondria. Moreover, CHK1 activation is required for the G2/M arrest and the generation of mitochondrial stress in response to RAD51 depletion. Together, our results indicate that nuclear DNA damage caused by RAD51 depletion may trigger mitochondria-originated redox dysregulation. Our findings suggest that a vicious cycle of nuclear DNA damage, mitochondrial accumulation and oxidative stress may contribute to the tumor-suppressive effects of RAD51 depletion or inhibition. RAD51 is overexpressed in ovarian cancer and is associated with poor prognosis. Depletion of RAD51 leads to increased mitochondrial superoxide production and oxidative DNA damage. Increased production of mitochondrial ROS requires CHK1-mediated G2/M arrest. mROS increase is independent of mtDNA.
Collapse
Affiliation(s)
- Limei Xu
- Key Laboratory of Experimental Teratology, Ministry of Education/Department of Molecular Medicine and Genetics, Shandong University School of Basic Medical Science, Jinan, Shandong, 250012, China
| | - Tingting Wu
- Key Laboratory of Experimental Teratology, Ministry of Education/Department of Molecular Medicine and Genetics, Shandong University School of Basic Medical Science, Jinan, Shandong, 250012, China
| | - Shihua Lu
- Key Laboratory of Experimental Teratology, Ministry of Education/Department of Molecular Medicine and Genetics, Shandong University School of Basic Medical Science, Jinan, Shandong, 250012, China
| | - Xiaohe Hao
- Key Laboratory of Experimental Teratology, Ministry of Education/Department of Molecular Medicine and Genetics, Shandong University School of Basic Medical Science, Jinan, Shandong, 250012, China
| | - Junchao Qin
- Department of Cell Biology, Shandong University School of Basic Medical Science, Jinan, Shandong, 250012, China
| | - Jing Wang
- Key Laboratory of Experimental Teratology, Ministry of Education/Department of Molecular Medicine and Genetics, Shandong University School of Basic Medical Science, Jinan, Shandong, 250012, China
| | - Xiyu Zhang
- Key Laboratory of Experimental Teratology, Ministry of Education/Department of Molecular Medicine and Genetics, Shandong University School of Basic Medical Science, Jinan, Shandong, 250012, China
| | - Qiao Liu
- Key Laboratory of Experimental Teratology, Ministry of Education/Department of Molecular Medicine and Genetics, Shandong University School of Basic Medical Science, Jinan, Shandong, 250012, China
| | - Beihua Kong
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Yaoqin Gong
- Key Laboratory of Experimental Teratology, Ministry of Education/Department of Molecular Medicine and Genetics, Shandong University School of Basic Medical Science, Jinan, Shandong, 250012, China
| | - Zhaojian Liu
- Department of Cell Biology, Shandong University School of Basic Medical Science, Jinan, Shandong, 250012, China
| | - Changshun Shao
- State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
36
|
Gong X, Li W, Dong L, Qu F. CircUBAP2 promotes SEMA6D expression to enhance the cisplatin resistance in osteosarcoma through sponging miR-506-3p by activating Wnt/β-catenin signaling pathway. J Mol Histol 2020; 51:329-340. [PMID: 32472335 PMCID: PMC7368871 DOI: 10.1007/s10735-020-09883-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 05/15/2020] [Indexed: 11/25/2022]
Abstract
The occurrence of chemo-resistance is an essential reason for the high morbidity of osteosarcoma (OS) patients. Circular RNAs (circRNAs) have been involved in the regulation of chemo-resistance in cancers. Semaphorins 6D (SEMA6D) is abnormally expressed in many cancers. However, the roles of circUBAP2 and SEMA6D in the chemo-resistance of OS are still unclear. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to detect the expression levels of circUBAP2, SEMA6D and microRNA-506-3p (miR-506-3p). The cisplatin resistance and proliferation of cells were evaluated by 3-(4, 5-dimethyl-2 thiazolyl)-2, 5-diphenyl-2-H-tetrazolium bromide assay. Western blot analysis was performed to measure the protein levels of Wnt/β-catenin signaling pathway biomarkers and SEMA6D. Also, the apoptosis, migration and invasion of cells were assessed by Flow cytometry and Transwell assays, respectively. Besides, Dual-luciferase reporter assay was used to verify the interaction between miR-506-3p and circUBAP2 or SEMA6D. We found that the expression levels of circUBAP2 and SEMA6D were increased in cisplatin-resistant OS tissues and cells. Knockdown of circUBAP2 inhibited the cisplatin resistance, silenced Wnt/β-catenin signaling pathway, hindered cell proliferation, migration and invasion, and promoted apoptosis in cisplatin-resistant OS cells, all of which could be reversed by overexpression of SEMA6D. MiR-506-3p could be sponged by circUBAP2 and could target SEMA6D. The suppression of miR-506-3p overexpression on the progression of OS cisplatin resistance could be reversed by SEMA6D overexpression, while miR-506-3p inhibitor also could invert the inhibitory effect of circUBAP2 silencing on the progression of OS cisplatin resistance. In conclusion, CircUBAP2 and SEMA6D played active roles in the progression of OS cisplatin resistance through miR-506-3p, which might provide some new ideas for studying the countermeasures of OS resistance.
Collapse
Affiliation(s)
| | | | - Lin Dong
- Department of Pharmacy, Yantai Affiliated Hospital of Binzhou Medical University, No. 717 Jinbu Avenue, Mouping District, Yantai, 264000, Shandong, China.
| | - Fangfei Qu
- Department of Special Inspection, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| |
Collapse
|
37
|
Zheng H, Zhang M, Ma S, Yang W, Xie S, Wang Y, Liu Y, Kai J, Ma Q, Lu R, Guo L. Identification of the key genes associated with chemotherapy sensitivity in ovarian cancer patients. Cancer Med 2020; 9:5200-5209. [PMID: 32441484 PMCID: PMC7367617 DOI: 10.1002/cam4.3122] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/17/2020] [Accepted: 04/25/2020] [Indexed: 12/17/2022] Open
Abstract
Background Ovarian cancer (OC) is the deadliest gynecological cancer. The absence of biomarkers in early detection and chemotherapy resistance is a principal cause of treatment failure in OC. Methods In this study, next generation sequencing (NGS) was used to sequence the mRNA of 44 OC patients including 14 chemotherapy insensitive and 18 sensitive patients. Differentially expressed genes (DEGs) from OC patients (compared with healthy controls) and chemotherapy sensitive patients (compared with chemotherapy insensitive patients) were identified by edgeR v3.12.0 in R v3.2.2, which were enriched using Gene Ontology (GO) database and Kyoto Encyclopedia of Genes and Genomes pathway (KEGG). The common DEGs in cancer occurring and chemotherapy sensitivity were further screened. Among them, genes participating in chemotherapy sensitivity associated pathways were regarded as chemotherapy sensitivity‐related key genes. Quantitative real‐time PCR (qPCR) and immunohistochemistry (IHC) were used to verify the expression of the key genes. Results We found 1588 DEGs between OC patients and healthy controls (HCs), which were mainly enriched in cell cycle pathway. Meanwhile, 249 DEGs were identified between chemotherapy sensitive and insensitive OC patients, which were mainly enriched in MAPK signaling pathway, ERBB signaling pathway, TNF signaling pathway, and IL‐17 signaling pathway. Thirty‐five DEGs were shared in chemotherapy sensitivity group and cancer occurring group. Among them, there are five genes (JUND, JUNB, MUC5B, NRG1, and NR4A1) participating in the above four chemotherapy sensitivity‐related pathways. It is remarkable that JUND is in the upstream of MUC5B in IL‐17 signaling pathway and their expressions were verified by qPCR and IHC. Conclusions The expression levels of the key genes related to chemotherapy sensitivity might be used as biomarkers to predict the treatment outcome and as a target to improve prognosis.
Collapse
Affiliation(s)
- Hui Zheng
- Department of Clinical LaboratoryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Meiqin Zhang
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
- Department of Gynecologic OncologyFudan University Shanghai Cancer CenterShanghaiChina
| | - Shuang Ma
- Genenexus Technology CorporationShanghaiChina
| | | | - Suhong Xie
- Department of Clinical LaboratoryFudan University Shanghai Cancer CenterShanghaiChina
| | - Yanchun Wang
- Department of Clinical LaboratoryFudan University Shanghai Cancer CenterShanghaiChina
| | - Yixuan Liu
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Jinyan Kai
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Qian Ma
- Department of Clinical LaboratoryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Renquan Lu
- Department of Clinical LaboratoryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Lin Guo
- Department of Clinical LaboratoryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| |
Collapse
|
38
|
Liang JJ, Wang JY, Zhang TJ, An GS, Ni JH, Li SY, Jia HT. MiR-509-3-5p-NONHSAT112228.2 Axis Regulates p21 and Suppresses Proliferation and Migration of Lung Cancer Cells. Curr Top Med Chem 2020; 20:835-846. [DOI: 10.2174/1568026620666200306102713] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 11/24/2019] [Accepted: 12/06/2019] [Indexed: 12/19/2022]
Abstract
Background:
Although the involvement of individual microRNA and lncRNA in the regulation
of p21 expression has largely been evidenced, less is known about the roles of functional interactions
between miRNAs and lncRNAs in p21 expression. Our previous work demonstrated that miR-509-
3-5p could block cancer cell growth.
Methods:
To gain an insight into the role of miR-509-3-5p in the regulation of p21 expression, we performed
in silico prediction and showed that miR-509-3-5p might target the NONHSAT112228.2, a
sense-overlapping lncRNA transcribed by a non-code gene overlapping with p21 gene. Mutation and
luciferase report analysis suggested that miR-509-3-5p could target NONHSAT112228.2, thereby blocking
its expression. Consistently, NONHSAT112228.2 expression was inversely correlated with both
miR-509-3-5p and p21 expression in cancer cells. Ectopic expression of miR-509-3-5p and knockdown
of NONHSAT112228.2 both promoted proliferation and migration of cancer cells.
Results:
Interestingly, high-expression of NONHSAT112228.2 accompanied by low-expression of p21
was observed in lung cancer tissues and associated with lower overall survival.
Conclusion:
Taken together, our study found a new regulatory pathway of p21, in which MiR-509-3-5p
functionally interacts with NONHSAT112228.2 to release p21 expression. MiR-509-3-5p—
NONHSAT112228.2 regulatory axis can inhibit the proliferation and migration of lung cancer cells.
Collapse
Affiliation(s)
- Jing-Jing Liang
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Jun-Yi Wang
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Tong-Jia Zhang
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Guo-Shun An
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Ju-Hua Ni
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Shu-Yan Li
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Hong-Ti Jia
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| |
Collapse
|
39
|
Ghaffari M, Asadi M, Shanehbandi D, Bornehdeli S, Sadeghzadeh M, Mohammad Reza Khani H, Ghasembaglou S. Aberrant Expression of miR-103, miR-184, miR-378, miR-497 and miR-506 in Tumor Tissue from Patients with Oral Squamous Cell Carcinoma Regulates the Clinical Picture of the Patients. Asian Pac J Cancer Prev 2020; 21:1311-1315. [PMID: 32458638 PMCID: PMC7541872 DOI: 10.31557/apjcp.2020.21.5.1311] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 03/01/2020] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND This study aimed to evaluate the expression patterns of miR-103, miR-184, miR-378, miR497 and in squamous cell carcinoma (SCC) of the tongue and to be compared with normal peripheral tissues. METHODS Tumor and marginal tissues were obtained from 50 patients with OSCC. After RNA extraction, expression level of miR-103, miR-184, miR-378, miR497, and miR506 was estimated using SYBR green master mix and real-time quantitative PCR. RESULTS It was observed that, there was no detectable difference in expression level of miR-103 between tumoral and marginal tissues. However, expression level of miR-184, and miR-378 showed significant increase in tumor tissue samples compared to marginal tissue samples. MiR-497 and miR-506 demonstrated considerable decrease in tumoral cells in comparison with peripheral tissues. Moreover, the expression level of miRNAs was associated with clinicopathological features of the patients. CONCLUSIONS Our data indicated that miR-184, miR-378, miR-497, and miR-506 can be used as a potential diagnostic and prognostic biomarker in OSCC. Nevertheless, more studies are needed to confirm this claim. .
Collapse
Affiliation(s)
- Maryam Ghaffari
- Tuberculosis and Lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Milad Asadi
- Tuberculosis and Lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Dariush Shanehbandi
- Tuberculosis and Lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Soghra Bornehdeli
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mahsa Sadeghzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | | - Shahram Ghasembaglou
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Medical Faculty, Tabriz University of Medical Science, Tabriz, Iran.
| |
Collapse
|
40
|
Drápela S, Bouchal J, Jolly MK, Culig Z, Souček K. ZEB1: A Critical Regulator of Cell Plasticity, DNA Damage Response, and Therapy Resistance. Front Mol Biosci 2020; 7:36. [PMID: 32266287 PMCID: PMC7096573 DOI: 10.3389/fmolb.2020.00036] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 02/14/2020] [Indexed: 12/29/2022] Open
Abstract
The predominant way in which conventional chemotherapy kills rapidly proliferating cancer cells is the induction of DNA damage. However, chemoresistance remains the main obstacle to therapy effectivity. An increasing number of studies suggest that epithelial-to-mesenchymal transition (EMT) represents a critical process affecting the sensitivity of cancer cells to chemotherapy. Zinc finger E-box binding homeobox 1 (ZEB1) is a prime element of a network of transcription factors controlling EMT and has been identified as an important molecule in the regulation of DNA damage, cancer cell differentiation, and metastasis. Recent studies have considered upregulation of ZEB1 as a potential modulator of chemoresistance. It has been hypothesized that cancer cells undergoing EMT acquire unique properties that resemble those of cancer stem cells (CSCs). These stem-like cells manifest enhanced DNA damage response (DDR) and DNA repair capacity, self-renewal, or chemoresistance. In contrast, functional experiments have shown that ZEB1 induces chemoresistance regardless of whether other EMT-related changes occur. ZEB1 has also been identified as an important regulator of DDR by the formation of a ZEB1/p300/PCAF complex and direct interaction with ATM kinase, which has been linked to radioresistance. Moreover, ATM can directly phosphorylate ZEB1 and enhance its stability. Downregulation of ZEB1 has also been shown to reduce the abundance of CHK1, an effector kinase of DDR activated by ATR, and to induce its ubiquitin-dependent degradation. In this perspective, we focus on the role of ZEB1 in the regulation of DDR and describe the mechanisms of ZEB1-dependent chemoresistance.
Collapse
Affiliation(s)
- Stanislav Drápela
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czechia.,International Clinical Research Center, Center for Biomolecular and Cellular Engineering, St. Anne's University Hospital in Brno, Brno, Czechia.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Jan Bouchal
- Department of Clinical and Molecular Pathology, Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Palacky University, Olomouc, Czechia
| | - Mohit Kumar Jolly
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Zoran Culig
- International Clinical Research Center, Center for Biomolecular and Cellular Engineering, St. Anne's University Hospital in Brno, Brno, Czechia.,Department of Urology, Experimental Urology, Innsbruck Medical University, Innsbruck, Austria
| | - Karel Souček
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czechia.,International Clinical Research Center, Center for Biomolecular and Cellular Engineering, St. Anne's University Hospital in Brno, Brno, Czechia.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| |
Collapse
|
41
|
Luo J, Dai X, Hu H, Chen J, Zhao L, Yang C, Sun J, Zhang L, Wang Q, Xu S, Xu Y, Liu N, Ying G, Wang P. Fluzoparib increases radiation sensitivity of non-small cell lung cancer (NSCLC) cells without BRCA1/2 mutation, a novel PARP1 inhibitor undergoing clinical trials. J Cancer Res Clin Oncol 2020; 146:721-737. [PMID: 31786739 DOI: 10.1007/s00432-019-03097-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 11/22/2019] [Indexed: 12/12/2022]
Abstract
PROPOSE Poly (ADP-ribose) polymerase 1 inhibitors were originally investigated as anti-cancer therapeutics with BRCA1/2 genes mutation. Here, we investigate the effectiveness of a novel PARP1 inhibitor fluzoparib, for enhancing the radiation sensitivity of NSCLC cells lacking BRCA1/2 mutation. METHODS We used MTS assays, western blotting, colony formation assays, immunofluorescence staining, and flow cytometry to evaluate the radiosensitization of NSCLC cells to fluzoparib and explore the underlying mechanisms in vitro. Through BRCA1 and RAD50 genes knockdown, we established dysfunctional homologous recombination (HR) DNA repair pathway models in NSCLC cells. We next investigated the radiosensitization effect of fluzoparib in vivo using human NSCLC xenograft models in mice. The expression of PARP1 and BRCA1 in human NSCLC tumor samples was measured by immunohistochemistry. Furthermore, we sequenced HR-related gene mutations and analyzed their frequencies in advanced NSCLC. RESULTS In vitro experiments in NSCLC cell lines along with in vivo experiments using an NSCLC xenograft mouse model demonstrated the radiosensitization effect of fluzoparib. The underlying mechanisms involved increased apoptosis, cell-cycle arrest, enhanced irradiation-induced DNA damage, and delayed DNA-damage repair. Immunohistochemical staining showed no correlation between the expression of PARP1 and BRCA1. Moreover, our sequencing results revealed high mutation frequencies for the BRCA1/2, CHEK2, ATR, and RAD50 genes. CONCLUSION The potential therapeutic value of fluzoparib for increasing the radiation sensitivity of NSCLC is well confirmed. Moreover, our findings of high mutation frequencies among HR genes suggest that PARP1 inhibition may be an effective treatment strategy for advanced non-small cell lung cancer patients.
Collapse
Affiliation(s)
- Jing Luo
- Laboratory of Cancer Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Xinchi Dai
- Laboratory of Cancer Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Hua Hu
- Pulmonary and Critical Care Medicine (PCCM), Shandong Chest Hospital Affiliated to Shandong University, Jinan, Shandong, 250013, China
| | - Jie Chen
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Lujun Zhao
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Changyong Yang
- Jiangsu Hengrui Medicine Co Ltd, Lianyungang, 222002, China
| | - Jifeng Sun
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Lianmin Zhang
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Qian Wang
- Laboratory of Cancer Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Shilei Xu
- Laboratory of Cancer Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Yue Xu
- Laboratory of Cancer Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Ningbo Liu
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
| | - Guoguang Ying
- Laboratory of Cancer Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
| | - Ping Wang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| |
Collapse
|
42
|
Taylor SJ, Arends MJ, Langdon SP. Inhibitors of the Fanconi anaemia pathway as potential antitumour agents for ovarian cancer. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2020; 1:26-52. [PMID: 36046263 PMCID: PMC9400734 DOI: 10.37349/etat.2020.00003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 12/18/2019] [Indexed: 11/30/2022] Open
Abstract
The Fanconi anaemia (FA) pathway is an important mechanism for cellular DNA damage repair, which functions to remove toxic DNA interstrand crosslinks. This is particularly relevant in the context of ovarian and other cancers which rely extensively on interstrand cross-link generating platinum chemotherapy as standard of care treatment. These cancers often respond well to initial treatment, but reoccur with resistant disease and upregulation of DNA damage repair pathways. The FA pathway is therefore of great interest as a target for therapies that aim to improve the efficacy of platinum chemotherapies, and reverse tumour resistance to these. In this review, we discuss recent advances in understanding the mechanism of interstrand cross-link repair by the FA pathway, and the potential of the component parts as targets for therapeutic agents. We then focus on the current state of play of inhibitor development, covering both the characterisation of broad spectrum inhibitors and high throughput screening approaches to identify novel small molecule inhibitors. We also consider synthetic lethality between the FA pathway and other DNA damage repair pathways as a therapeutic approach.
Collapse
Affiliation(s)
- Sarah J Taylor
- Cancer Research UK Edinburgh Centre and Edinburgh Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XU Edinburgh, UK
| | - Mark J Arends
- Cancer Research UK Edinburgh Centre and Edinburgh Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XU Edinburgh, UK
| | - Simon P Langdon
- Cancer Research UK Edinburgh Centre and Edinburgh Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XU Edinburgh, UK
| |
Collapse
|
43
|
Sun C, Cao W, Qiu C, Li C, Dongol S, Zhang Z, Dong R, Song K, Yang X, Zhang Q, Kong B. MiR-509-3 augments the synthetic lethality of PARPi by regulating HR repair in PDX model of HGSOC. J Hematol Oncol 2020; 13:9. [PMID: 32005272 PMCID: PMC6995078 DOI: 10.1186/s13045-020-0844-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 01/15/2020] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND PARP inhibitors have been the most promising target drugs with widely proven benefits among ovarian cancer patients. Although platinum-response, HR-related genes, or HRD genomic scar detection are acceptably used in assessment of Olaparib response, there are still evident limitations in the present approaches. Therefore, we aim to investigate more accurate approaches to predict Olaparib sensitivity and effective synergistic treatment strategies. METHODS We probed two databases (TCGA and Qilu Hospital) in order to quest novel miRNAs associated with platinum-sensitivity or HR-related genes. Cellular experiments in vitro or in vivo and PDX models were utilized to validate their role in tumor suppression and Olaparib sensitizing. Furthermore, HR gene mutation was analyzed through WES to explore the relation between HR gene mutation and Olaparib response. RESULTS High miR-509-3 expression indicated better response to platinum and longer progression-free and overall survival in two independent ovarian cancer patient cohorts (high vs. low miR-509-3 expression; PFS: TCGA P < 0.05, Qilu P < 0.05; OS: TCGA P < 0.05, Qilu P < 0.01). MiR-509-3 could impair the proliferation, migration, and invasion ability but enhance the sensitivity to Olaparib of ovarian cancer cell in vitro and in vivo by directly targeting HMGA2 and RAD51. In two PDX cases (PDX1 and PDX9), miR-509-3 could significantly increase the sensitivity to Olaparib along with the decrease of RAD51 positive rate (mean tumor weight NC + Olaparib vs. miR-509 + Olaparib; PDX1 P < 0.05, PDX9 P < 0.05). Additionally, in PDX8, miR-509-3 treatment dramatically reversed the Olaparib insensitivity (P < 0.05) by downregulating RAD51 expression. RAD51 functional detection revealed that all Olaparib sensitive cases exhibited low RAD51 positive rate (lesser than 50%) in treated groups. Furthermore, among the four HR gene mutation patients, three harbored HR core gene mutation and were sensitive to Olaparib while the remaining one with non-HR core gene mutation did not respond well to Olaparib. CONCLUSIONS MiR-509-3 can sensitize ovarian cancer cells to Olaparib by impeding HR, which makes it a potential target in PARPi synergistic treatment. HR core gene analysis and RAD51 functional detection are prospectively feasible in prediction of PARPi response.
Collapse
Affiliation(s)
- Chenggong Sun
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, People's Republic of China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, People's Republic of China
| | - Wenyu Cao
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, People's Republic of China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, People's Republic of China
| | - Chunping Qiu
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, People's Republic of China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, People's Republic of China
| | - Chengcheng Li
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, People's Republic of China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, People's Republic of China
| | - Samina Dongol
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, People's Republic of China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, People's Republic of China
| | - Zhiwei Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, People's Republic of China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, People's Republic of China
| | - Ruifen Dong
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, People's Republic of China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, People's Republic of China
| | - Kun Song
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, People's Republic of China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, People's Republic of China
| | - Xingsheng Yang
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, People's Republic of China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, People's Republic of China
| | - Qing Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, People's Republic of China. .,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, People's Republic of China.
| | - Beihua Kong
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, People's Republic of China. .,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, People's Republic of China.
| |
Collapse
|
44
|
Role of Rad51 and DNA repair in cancer: A molecular perspective. Pharmacol Ther 2020; 208:107492. [PMID: 32001312 DOI: 10.1016/j.pharmthera.2020.107492] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/13/2020] [Accepted: 01/22/2020] [Indexed: 12/24/2022]
Abstract
The maintenance of genome integrity is essential for any organism survival and for the inheritance of traits to offspring. To the purpose, cells have developed a complex DNA repair system to defend the genetic information against both endogenous and exogenous sources of damage. Accordingly, multiple repair pathways can be aroused from the diverse forms of DNA lesions, which can be effective per se or via crosstalk with others to complete the whole DNA repair process. Deficiencies in DNA healing resulting in faulty repair and/or prolonged DNA damage can lead to genes mutations, chromosome rearrangements, genomic instability, and finally carcinogenesis and/or cancer progression. Although it might seem paradoxical, at the same time such defects in DNA repair pathways may have therapeutic implications for potential clinical practice. Here we provide an overview of the main DNA repair pathways, with special focus on the role played by homologous repair and the RAD51 recombinase protein in the cellular DNA damage response. We next discuss the recombinase structure and function per se and in combination with all its principal mediators and regulators. Finally, we conclude with an analysis of the manifold roles that RAD51 plays in carcinogenesis, cancer progression and anticancer drug resistance, and conclude this work with a survey of the most promising therapeutic strategies aimed at targeting RAD51 in experimental oncology.
Collapse
|
45
|
Wambecke A, Ahmad M, Lambert B, Joly F, Poulain L, Denoyelle C, Meryet-Figuiere M. The influence of long non-coding RNAs on the response to chemotherapy in ovarian cancer. Gynecol Oncol 2019; 156:726-733. [PMID: 31883617 DOI: 10.1016/j.ygyno.2019.12.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/05/2019] [Accepted: 12/16/2019] [Indexed: 12/14/2022]
Abstract
With 240,000 new cases and 152,000 deaths per year, ovarian cancer is the leading cause of death from gynecologic malignancies. Late diagnosis because of asymptomatic development in early stages and resistance to existing treatments are the major causes of therapeutic failure in ovarian cancer. The recent discovery of tens of thousands of long non-coding RNAs and their action as oncogenes or tumor suppressors in pathways matching all the hallmarks of cancer in most - if not all - malignancies have attracted attention of the scientific community. A growing number of studies have implicated lncRNAs in diverse aspects of ovarian carcinoma biology. We present lncRNAs which have been involved in response to the different drugs currently used for the treatment of ovarian cancers, from first-line platinum salts and taxanes to the newly available PARP inhibitors. The data already available supports the potential use of several lncRNAs, alone or in combination with other molecules, as potential biomarkers for the prediction of response to treatment. Understanding the determinants of their action might reveal new potential therapeutic targets.
Collapse
Affiliation(s)
- Anaïs Wambecke
- Normandie Univ, UNICAEN, Inserm U1086 ANTICIPE (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Caen, France; Cancer Centre François Baclesse, UNICANCER, Caen, France
| | - Mohammad Ahmad
- Normandie Univ, UNICAEN, Inserm U1086 ANTICIPE (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Caen, France; Cancer Centre François Baclesse, UNICANCER, Caen, France
| | - Bernard Lambert
- Normandie Univ, UNICAEN, Inserm U1086 ANTICIPE (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Caen, France; Cancer Centre François Baclesse, UNICANCER, Caen, France; CNRS, Normandy Regional Delegation, Caen, France
| | - Florence Joly
- Normandie Univ, UNICAEN, Inserm U1086 ANTICIPE (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Caen, France; Cancer Centre François Baclesse, UNICANCER, Caen, France
| | - Laurent Poulain
- Normandie Univ, UNICAEN, Inserm U1086 ANTICIPE (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Caen, France; Cancer Centre François Baclesse, UNICANCER, Caen, France
| | - Christophe Denoyelle
- Normandie Univ, UNICAEN, Inserm U1086 ANTICIPE (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Caen, France; Cancer Centre François Baclesse, UNICANCER, Caen, France
| | - Matthieu Meryet-Figuiere
- Normandie Univ, UNICAEN, Inserm U1086 ANTICIPE (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Caen, France; Cancer Centre François Baclesse, UNICANCER, Caen, France.
| |
Collapse
|
46
|
Wang J, Du Y, Deng J, Wang X, Long F, He J. MicroRNA-506 Is Involved in Regulation of the Occurrence of Lipopolysaccharides (LPS)-Induced Pulpitis by Sirtuin 1 (SIRT1). Med Sci Monit 2019; 25:10008-10015. [PMID: 31877121 PMCID: PMC6944165 DOI: 10.12659/msm.918172] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Toothache often occurs with pulpitis. Lipopolysaccharide (LPS) is produced by gram-negative bacteria, and its accumulation is related to clinical symptoms of pain. MicroRNAs (miRNAs) display anti-inflammatory potential due to their direct regulation of cellular protein expression, which can promote inflammatory changes in dental pulp tissues. However, the mechanism of LPS-induced pulpitis is still unclear. Material/Methods In this study, dental pulp stem cells (DPSCs) were separated and cultured from rat dental pulp tissues; then, LPS was administered to induce inflammation and activate the TLR4 pathway. Results It was found that miR-506 was upregulated following LPS treatment in DPSCs. The inhibition of miR-506 in LPS-treated DPSCs led to attenuated inflammation and deactivation of the TLR4 pathway. Furthermore, the bioinformatic analysis and dual-luciferase reporter gene assay indicated that miR-506 could target the 3′-UTR of sirtuin 1 (SIRT1). Additionally, SIRT1 decreased in LPS-treated DPSCs, and miR-506 transfection resulted in SIRT1 upregulation. SIRT1 overexpression showed a similar inhibitory effect as that of miR-506 downregulation on inflammation and TLR4 activation in DPSCs. Conclusions In brief, miR-506 can protect dental pulp in LPS-induced inflammation by inhibiting the SIRT1-mediated TLR4 pathway.
Collapse
Affiliation(s)
- Jun Wang
- Stomatological Center, Gansu Provincial Hospital, Lanzhou, Gansu, China (mainland)
| | - Yi Du
- Department of Nursing, The First Hospital of Lanzhou University, Lanzhou, Gansu, China (mainland)
| | - Junhong Deng
- Department of Stomatology, The Second People's Hospital of Lanzhou City, Lanzhou, Gansu, China (mainland)
| | - Xin Wang
- Departments of Health and Social Care, The Second People's Hospital of Lanzhou City, Lanzhou, Gansu, China (mainland)
| | - Fei Long
- Stomatological Center, Gansu Provincial Hospital, Lanzhou, Gansu, China (mainland)
| | - Jianmin He
- Stomatological Center, Gansu Provincial Hospital, Lanzhou, Gansu, China (mainland)
| |
Collapse
|
47
|
Xiao M, Guo J, Xie L, Yang C, Gong L, Wang Z, Cai J. Let-7e Suppresses DNA Damage Repair and Sensitizes Ovarian Cancer to Cisplatin through Targeting PARP1. Mol Cancer Res 2019; 18:436-447. [PMID: 31722968 DOI: 10.1158/1541-7786.mcr-18-1369] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 05/26/2019] [Accepted: 11/07/2019] [Indexed: 11/16/2022]
Abstract
Increased DNA damage repair is one of the mechanisms implicated in cisplatin resistance. Our previous study indicated that the deregulation of let-7e promoted cisplatin resistance and that let-7e could suppress DNA double-strand break repair in ovarian cancer. In this study, we further characterized the role of let-7e in DNA damage repair and cisplatin resistance in ovarian cancer, and investigated the underlying mechanisms. The alkaline and neutral comet assay indicated that let-7e impeded both DNA single- and double-strand break repairs through downregulating its target gene PARP1. In vitro and in vivo experiments provided evidence that the let-7e-PARP1-DNA repair axis was involved in the modulation of cisplatin sensitivity in ovarian cancer. Contrary to let-7e, PARP1 was overexpressed in cisplatin-resistant ovarian cancer tissues, and patients with high PARP1 expression exhibited poor progression-free survival (PFS) and overall survival (OS). Multivariate logistic and Cox regression analyses showed that let-7e and FIGO stage were independent prognostic factors for PFS and OS, whereas let-7e and PARP1 were able to independently predict chemotherapy response. Taken together, our results indicated that low expression of let-7e promoted DNA single- and double-strand break repairs and subsequently contributed to cisplatin resistance by relieving the suppression on PARP1 in ovarian cancer. IMPLICATIONS: Targeting the let-7e-PARP1-DNA repair axis might be an effective strategy for the treatment of chemoresistant ovarian cancer.
Collapse
Affiliation(s)
- Man Xiao
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianfeng Guo
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lisha Xie
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chun Yang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lanqing Gong
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zehua Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jing Cai
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
48
|
Patterson EL, Saski CA, Sloan DB, Tranel PJ, Westra P, Gaines TA. The Draft Genome of Kochia scoparia and the Mechanism of Glyphosate Resistance via Transposon-Mediated EPSPS Tandem Gene Duplication. Genome Biol Evol 2019; 11:2927-2940. [PMID: 31518388 PMCID: PMC6808082 DOI: 10.1093/gbe/evz198] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2019] [Indexed: 12/14/2022] Open
Abstract
Increased copy number of the 5-enolpyruvylshikimate-3-phosphate synthase (EPSPS) gene confers resistance to glyphosate, the world's most-used herbicide. There are typically three to eight EPSPS copies arranged in tandem in glyphosate-resistant populations of the weed kochia (Kochia scoparia). Here, we report a draft genome assembly from a glyphosate-susceptible kochia individual. Additionally, we assembled the EPSPS locus from a glyphosate-resistant kochia plant by sequencing select bacterial artificial chromosomes from a kochia bacterial artificial chromosome library. Comparing the resistant and susceptible EPSPS locus allowed us to reconstruct the history of duplication in the structurally complex EPSPS locus and uncover the genes that are coduplicated with EPSPS, several of which have a corresponding change in transcription. The comparison between the susceptible and resistant assemblies revealed two dominant repeat types. Additionally, we discovered a mobile genetic element with a FHY3/FAR1-like gene predicted in its sequence that is associated with the duplicated EPSPS gene copies in the resistant line. We present a hypothetical model based on unequal crossing over that implicates this mobile element as responsible for the origin of the EPSPS gene duplication event and the evolution of herbicide resistance in this system. These findings add to our understanding of stress resistance evolution and provide an example of rapid resistance evolution to high levels of environmental stress.
Collapse
Affiliation(s)
- Eric L Patterson
- Department of Bioagricultural Sciences and Pest Management, Colorado State University
- Department of Genetics and Biochemistry, Clemson University
| | | | | | | | - Philip Westra
- Department of Bioagricultural Sciences and Pest Management, Colorado State University
| | - Todd A Gaines
- Department of Bioagricultural Sciences and Pest Management, Colorado State University
| |
Collapse
|
49
|
Singh A, Gupta S, Sachan M. Epigenetic Biomarkers in the Management of Ovarian Cancer: Current Prospectives. Front Cell Dev Biol 2019; 7:182. [PMID: 31608277 PMCID: PMC6761254 DOI: 10.3389/fcell.2019.00182] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 08/19/2019] [Indexed: 12/15/2022] Open
Abstract
Ovarian cancer (OC) causes significant morbidity and mortality as neither detection nor screening of OC is currently feasible at an early stage. Difficulty to promptly diagnose OC in its early stage remains challenging due to non-specific symptoms in the early-stage of the disease, their presentation at an advanced stage and poor survival. Therefore, improved detection methods are urgently needed. In this article, we summarize the potential clinical utility of epigenetic signatures like DNA methylation, histone modifications, and microRNA dysregulation, which play important role in ovarian carcinogenesis and discuss its application in development of diagnostic, prognostic, and predictive biomarkers. Molecular characterization of epigenetic modification (methylation) in circulating cell free tumor DNA in body fluids offers novel, non-invasive approach for identification of potential promising cancer biomarkers, which can be performed at multiple time points and probably better reflects the prevailing molecular profile of cancer. Current status of epigenetic research in diagnosis of early OC and its management are discussed here with main focus on potential diagnostic biomarkers in tissue and body fluids. Rapid and point of care diagnostic applications of DNA methylation in liquid biopsy has been precluded as a result of cumbersome sample preparation with complicated conventional methods of isolation. New technologies which allow rapid identification of methylation signatures directly from blood will facilitate sample-to answer solutions thereby enabling next-generation point of care molecular diagnostics. To date, not a single epigenetic biomarker which could accurately detect ovarian cancer at an early stage in either tissue or body fluid has been reported. Taken together, the methodological drawbacks, heterogeneity associated with ovarian cancer and non-validation of the clinical utility of reported potential biomarkers in larger ovarian cancer populations has impeded the transition of epigenetic biomarkers from lab to clinical settings. Until addressed, clinical implementation as a diagnostic measure is a far way to go.
Collapse
Affiliation(s)
- Alka Singh
- Department of Biotechnology, Motilal Nehru National Institute of Technology, Allahabad, India
| | - Sameer Gupta
- Department of Surgical Oncology, King George Medical University, Lucknow, India
| | - Manisha Sachan
- Department of Biotechnology, Motilal Nehru National Institute of Technology, Allahabad, India
| |
Collapse
|
50
|
Zeng X, Baba T, Hamanishi J, Matsumura N, Kharma B, Mise Y, Abiko K, Yamaguchi K, Horikawa N, Hunstman DG, Mulati K, Kitamura S, Taki M, Murakami R, Hosoe Y, Mandai M. Phosphorylation of STAT1 serine 727 enhances platinum resistance in uterine serous carcinoma. Int J Cancer 2019; 145:1635-1647. [PMID: 31228268 DOI: 10.1002/ijc.32501] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 05/12/2019] [Accepted: 05/24/2019] [Indexed: 11/07/2022]
Abstract
Uterine serous carcinoma (USC) is a highly aggressive histological subtype of endometrial cancers harboring highly metastatic and chemoresistant features. Our previous study showed that STAT1 is highly expressed in USC and acts as a key molecule that is positively correlated with tumor progression, but it remains unclear whether STAT1 is relevant to the malicious chemorefractory nature of USC. In the present study, we investigated the regulatory role of STAT1 toward platinum-cytotoxicity in USC. STAT1 suppression sensitized USC cells to increase cisplatin-mediated apoptosis (p < 0.001). Furthermore, phosphorylation of STAT1 was prominently observed on serine-727 (pSTAT1-Ser727), but not on tyrosine-701, in the nucleus of USC cells treated with cisplatin. Mechanistically, the inhibition of pSTAT1-Ser727 by dominant-negative plasmid elevated cisplatin-mediated apoptosis by increasing intracellular accumulation of cisplatin through upregulation of CTR1 expression. TBB has an inhibitory effect on casein kinase 2 (CK2), which phosphorylate STAT1 at serine residues. Sequential treatment with TBB and cisplatin on USC cells greatly reduced nuclear pSTAT1-Ser727, enhanced intracellular accumulation of cisplatin, and subsequently increased apoptosis. Tumor load was significantly reduced by combination therapy of TBB and cisplatin in in vivo xenograft models (p < 0.001). Our results collectively suggest that pSTAT1-Ser727 may play a key role in platinum resistance as well as tumor progression in USC. Thus, targeting the STAT1 pathway via CK2 inhibitor can be a novel method for attenuating the chemorefractory nature of USC.
Collapse
Affiliation(s)
- Xiang Zeng
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tsukasa Baba
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, Iwate, Japan
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Noriomi Matsumura
- Department of Obstetrics and Gynecology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Budiman Kharma
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuka Mise
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kaoru Abiko
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ken Yamaguchi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Naoki Horikawa
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - David G Hunstman
- Department of Pathology and Laboratory Medicine, University of British Columbia, British Columbia Cancer Agency, Vancouver, BC, Canada.,Genetic Pathology Evaluation Centre, Vancouver General Hospital, Vancouver, BC, Canada
| | - Kumuluzi Mulati
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Sachiko Kitamura
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mana Taki
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryusuke Murakami
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuko Hosoe
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|